Language selection

Search

Patent 2386759 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 2386759
(54) English Title: THERAPEUTIC USES OF PEROXOMETALLIC COMPOUNDS
(54) French Title: USAGES THERAPEUTIQUES DES COMPOSES PEROXOMETALLIQUES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 33/24 (2006.01)
  • A61K 31/555 (2006.01)
  • A61K 39/008 (2006.01)
(72) Inventors :
  • FAURE, ROBERT (Canada)
  • SAVARD, PIERRE (Canada)
  • DOILLON, CHARLES (Canada)
  • BATTISTINI, BRUNO JOSEPH (Canada)
  • OLIVIER, MARTIN (Canada)
  • POSNER, BARRY (Canada)
(73) Owners :
  • UNIVERSITE LAVAL (Canada)
  • MCGILL UNIVERSITY (Canada)
(71) Applicants :
  • UNIVERSITE LAVAL (Canada)
  • MCGILL UNIVERSITY (Canada)
(74) Agent: GOUDREAU GAGE DUBUC
(74) Associate agent:
(45) Issued:
(22) Filed Date: 2002-05-17
(41) Open to Public Inspection: 2003-11-17
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): No

(30) Application Priority Data: None

Abstracts

English Abstract





The present invention relates to the use of peroxometallic compounds, such
peroxovanadium compounds, to prevent angiogenesis, restenosis and the
production of endothelins, as immunomodulators and as antitumorigenic
agents. Peroxometallic compounds are preferred since they are more
potent, and less toxic, than their "oxo" counterparts, as exemplified by
specific peroxovanadium compounds. Anti-angiogenic activity was verified in
vitro against human umbilical vascular endothelial cells (HUVECs) as well as
ex ovo using the chicken chorioallantoic assay membrane and in the rat
aortic ring model and a Matrigel assay in vivo. Peroxovanadium compounds
also decrease basal levels and inhibit the increase in plasma endothelins
occurring following insulin induction in rats. It is proposed that
peroxovanadium compounds are therapeutically-active anti-angiogenics and
useful in preventing vascular restenosis by acting, inter alia, by inhibiting
one
or several protein tyrosine phosphatases (PTPs) involved in the proliferation,
differentiation and migration of cells or the secretion of peptides (such
endothelins and immunomodulators), or both. These compounds have also
been found to be suitable as antitumorigenic agents in the treatment of
cancer, such as breast cancer and prostate cancer.


Claims

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





56

WHAT IS CLAIMED IS:

1. A method for preventing or arresting angiogenesis in an animal
comprising administering a therapeutically effective amount of a
compound of the following formula to prevent said angiogenesis

Image

wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,

Y is oxygen or hydroxyl,

Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

2. A method as defined in claim 1, wherein said angiogenesis is related
to arthritis, psoriasis, a disorder of the eye or a condition characterized
by the development of solid tumors.

3. A composition for preventing or arresting in an animal comprising a
pharmaceutically-acceptable excipient and a therapeutically effective
amount of a compound of the following formula to prevent said
angiogenesis





57

Image

wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

4. A composition as defined in claim 3, wherein said angiogenesis is
related to arthritis, psoriasis, a disorder of the eye or a condition
characterized by the development of solid tumors.

5. A device comprising a composition as defined in claim 3.

6. A method for preventing or lessening endothelin production in an
animal by inhibiting protein tyrosine phosphatases (PTPs) comprising
administering a therapeutically effective amount of a compound of the
following formula:

Image




58

wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

7. A composition for preventing or lessening endothelin production in an
animal by inhibiting protein tyrosine phosphatases (PTPs) comprising
a pharmaceutically-acceptable excipient and a therapeutically effective
amount of a compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.
8. A method for preventing restenosis following angioplasty in an animal
comprising administering a therapeutically effective amount of a



59

compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

9. A composition for preventing restenosis following angioplasty in an
animal comprising a pharmaceutically-acceptable excipient and a
therapeutically effective amount of a compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,



60

Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

10. A device comprising a composition as defined in claim 9.

11. A device as defined in claim 10, wherein said device is a stent.

12. A method for modulating inflammation and secretion of inflammatory
molecules in an animal comprising administering a therapeutically
effective amount of a compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

13. A composition for modulating inflammation and secretion of
inflammatory molecules in an animal comprising a pharmaceutically-
acceptable excipient and a therapeutically effective amount of a
compound of the following formula:



61

Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

14. A device comprising a composition as defined in claim 13.

15. A method for inducing the activation of cytokines and chemokines in an
animal comprising administering a therapeutically effective amount of a
compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,



62~

Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

16. A method as defined in claim 15, wherein said cytokines are IL-12,
IFN-.gamma., IL-1.alpha. or IL-1.beta. and said chemokines are RANTES, MIP-
1.alpha., MIP-
1.beta., MIP-2, IP-10 or MCP-1.

17. A composition for inducing the activation of cytokines and chemokines
in an animal comprising a pharmaceutically-acceptable excipient and a
therapeutically effective amount of a compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,~~
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

18. A composition as defined in claim 17, wherein said cytokines are IL-
12, IFN-.gamma., IL-1.alpha. or IL-1.beta. and said chemokines are RANTES, MIP-
1.alpha.,
MIP-1.beta., MIP-2, IP-10 or MCP-1.



63

19. An adjuvant having the following formula:
Image
wherein ~T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

20. A vaccine comprising an adjuvant as defined in claim 19.

21. A vaccine as defined in claim 20, wherein said vaccine is against
Leishmania parasite or other pathogens.

22. A method for preventing or arresting tumor growth in an animal
comprising administering a therapeutically effective amount of a
compound of the following formula:


64
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

23. ~A composition for preventing or arresting tumor growth in an animal
comprising a pharmaceutically-acceptable excipient and a
therapeutically effective amount of a compound of the following
formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,



65

Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

24. ~A device comprising a composition as defined in claim 23.

25. ~A method for treating cancer in an animal comprising administering a
therapeutically effective amount of a compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

26. A composition for treating cancer in an animal comprising a
pharmaceutically-acceptable excipient and a therapeutically effective
amount of a compound of the following formula:



66

Image~
wherein ~T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

27. A device comprising a composition as defined in claim 26.

28. A method as defined in claim 25, wherein said cancer is breast cancer or
prostate cancer.

29. A composition as defined in claim 26, wherein said cancer is breast
cancer or prostate cancer.

30. A vaccine against cancer comprising a compound of the following
formula:
Image



67

wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

31. ~A method for activating leucocytes in an animal comprising
administering to said animal leucocytes that have been treated in vitro
with a compound of the following formula:
Image
wherein T is a transition metal selected from the group consisting
of vanadium, molybdenum, tungsten, titanium, nobium
and tantalum,
Y is oxygen or hydroxyl,
Z and Z' are independently selected from oxygen and~
peroxide and at least one of them is peroxide, and
L or L' are any group which can donate one electron
pair.

32.A method as defined in claim 31 for use in the treatment of cancer.

33.A method as defined in claim 1, wherein T is vanadium.

34.A method as defined in claim 1, wherein T is molybdenum.

35.A method as defined in claim 1, wherein T is tungsten.



68
36.A method as defined in claim 1, wherein Z is oxygen and Z' is peroxide.
37.A method as defined in claim 1, wherein Z and Z' are peroxide.
38.A composition as defined in claim 3, wherein T is vanadium.
39.A composition as defined in claim 3, wherein T is molybdenum.
40.A composition as defined in claim 3, wherein T is tungsten.
41.A composition as defined in claim 3, wherein Z is oxygen and Z' is
peroxide.
42.A composition as defined in claim 3, wherein Z and Z' are peroxide.
43.A method as defined in claim 8, wherein T is vanadium.
44.A method as defined in claim 8, wherein T is molybdenum.
45.A method as defined in claim 8, wherein T is tungsten.
46.A method as defined in claim 8, wherein Z is oxygen and Z' is peroxide.
47.A method as defined in claim 8, wherein Z and Z' are peroxide.
48.A composition as defined in claim 9, wherein T is vanadium.
49.A composition as defined in claim 9, wherein T is molybdenum.
50.A composition as defined in claim 9, wherein T is tungsten.
51.A composition as defined in claim 9, wherein Z is oxygen and Z' is
peroxide.
52.A composition as defined in claim 9, wherein Z and Z' are peroxide.
53.A device comprising a composition as defined in claim 48.
54.A device comprising a composition as defined in claim 49.
55.A device comprising a composition as defined in claim 50.
56.A device comprising a composition as defined in claim 51.
57.A device comprising a composition as defined in claim 52.
58.A device as defined in claim 53, wherein said device is a stent.
59.A device as defined in claim 54, wherein said device is a stent.
60.A device as defined in claim 55, wherein said device is a stent.
61.A device as defined in claim 56, wherein said device is a stent.
62.A device as defined in claim 57, wherein said device is a stent.
63.An adjuvant as defined in claim 19, wherein T is vanadium.
64.An adjuvant as defined in claim 19, wherein T is molybdenum.
65.An adjuvant as defined in claim 19, wherein T is tungsten.



69
66.An adjuvant as defined in claim 19, wherein Z is oxygen and Z' is
peroxide.
67.An adjuvant as defined in claim 19, wherein Z and Z' are peroxide.
68.A vaccine comprising an adjuvant as defined in claim 63.
69.A vaccine comprising an adjuvant as defined in claim 64.
70.A vaccine comprising an adjuvant as defined in claim 65.
71.A vaccine comprising an adjuvant as defined in claim 66.
72.A vaccine comprising an adjuvant as defined in claim 67.
73.A method as defined in claim 22, wherein T is vanadium.
74.A method as defined in claim 22, wherein T is molybdenum.
75.A method as defined in claim 22, wherein T is tungsten.
76.A method as defined in claim 22, wherein Z is oxygen and Z' is peroxide.
77.A method as defined in claim 22, wherein Z and Z' are peroxide.
78.A composition as defined in claim 23, wherein T is vanadium.
79.A composition as defined in claim 23, wherein T is molybdenum.
80.A composition as defined in claim 23, wherein T is tungsten.
81.A composition as defined in claim 23, wherein Z is oxygen and Z' is
peroxide.
82.A composition as defined in claim 23, wherein Z and Z' are peroxide.
83.A device comprising a composition as defined in claim 78.
84.A device comprising a composition as defined in claim 79.
85.A device comprising a composition as defined in claim 80.
86.A device comprising a composition as defined in claim 81.
87.A device comprising a composition as defined in claim 82.
88.A method as defined in claim 25, wherein T is vanadium.
89.A method as defined in claim 25, wherein T is molybdenum.
90.A method as defined in claim 25, wherein T is tungsten.
91.A method as defined in claim 25, wherein Z is oxygen and Z' is peroxide.
92.A method as defined in claim 25, wherein Z and Z' are peroxide.
93.A composition as defined in claim 26, wherein T is vanadium.
94.A composition as defined in claim 26, wherein T is molybdenum.
95.A composition as defined in claim 26, wherein T is tungsten.




70

96.A composition as defined in claim 26, wherein Z is oxygen and Z' is
peroxide.

97.A composition as defined in claim 26, wherein Z and Z' are peroxide.

98.A device comprising a composition as defined in claim 93.

99.A device comprising a composition as defined in claim 94.

100. A device comprising a composition as defined in claim 95.

101. A device comprising a composition as defined in claim 96.

102. A device comprising a composition as defined in claim 97.

103. A vaccine as defined in claim 30, wherein T is vanadium.

104. A vaccine as defined in claim 30, wherein T is molybdenum.

105. A vaccine as defined in claim 30, wherein T is tungsten.

106. A vaccine as defined in claim 30, wherein Z is oxygen and Z' is
peroxide.

107. A vaccine as defined in claim 30, wherein Z and Z' are peroxide.

108. A method as defined in claim 31, wherein T is vanadium.

109. A method as defined in claim 31, wherein T is molybdenum.

110. A method as defined in claim 31, wherein T is tungsten.

111. A method as defined in claim 31, wherein Z is oxygen and Z' is
peroxide.

112. A method as defined in claim 31, wherein Z and Z' are peroxide.

113. A method as defined in claim 32, wherein T is vanadium.

114. A method as defined in claim 32, wherein T is molybdenum.

115. A method as defined in claim 32, wherein T is tungsten.

116. A method as defined in claim 32, wherein Z is oxygen and Z' is
peroxide.

117. A method as defined in claim 32, wherein Z and Z' are peroxide.

118. A method as defined in claim 1, wherein said compound is bpV(phen),
bpV(pic) or bpV(bipy).

119. A composition as defined in claim 3, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).

120. A method as defined in claim 8, wherein said compound is bpV(phen),
bpV(pic) or bpV(bipy).



71
121. A composition as defined in claim 9, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
122. A device comprising a composition as defined in claim 121.
123. A device as defined in claim 122, wherein said device is a stem.
124. An adjuvant as defined in claim 19, wherein said adjuvant is
bpV(phen), bpV(pic) or bpV(bipy).
125. A vaccine comprising an adjuvant as defined in claim 124.
126. A vaccine as defined in claim 125, wherein said vaccine is against
Leishmania parasite or other pathogens.
127. A method as defined in claim 22, wherein wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
128. A composition as defined in claim 23, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
129. A device comprising a composition as defined in claim 128.
130. A method as defined in claim 25, wherein wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
131. A composition as defined in claim 26, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
132. A device comprising a composition as defined in claim 131.
133. A vaccine as defined in claim 30, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
134. A method as defined in claim 31, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).
135. A method as defined in claim 32, wherein said compound is
bpV(phen), bpV(pic) or bpV(bipy).

Description

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


CA 02386759 2002-05-17
1
TITLE OF THE INVENTION
Therapeutic Uses of Peroxometallic Compounds
FIELD OF THE INVENTION
This invention relates to the use of peroxometallic compounds, such as
potassium bisperoxo(1,10-phenanthroline)oxovanadate (bpV(phen)],
potassium bisperoxo(pyridine-2-carboxylato)oxovanadate [bpV(pic)] and
potassium bisperoxo(2,2'-bipyridyl) oxovanadate [bpV(bipy)], for preventing
angiogenesis, restenosis and the production of endothelins, and as adjuvants
for vaccination. The compounds of the present invention are also suitable as
antitumorigenic agents; for example, experimental results reveal that they are
efficacious in the treatment of breast cancer and prostate cancer.
BACKGROUND OF THE INVENTION
A) PEROXOVANADIUM COMPOUNDS:
Synthetic peroxovanadium (pV) compounds are structurally versatile
molecules which are potent inhibitors of phosphotyrosyl phosphatases
(PTPs) (1 ). These compounds contain one oxo ligand, one or two peroxo
groups, one ancillary ligand, all coordinated to vanadium. They are stable in
aqueous solution at physiological pH when shielded from light. pVs are
cytostatic agents, unlike many other antitumor molecules. Their mode of
. action lies in the modulation of the activity of cellular transduction
pathways
involved in the progression of pathological conditions. Their effects are
transitory and disappear within a few days after administration(2).
Phosphotyrosine phosphatases (PTPs) are enzymes which remove
phosphates from tyrosine residues of proteins. They are involved in several
cell functions regulating proliferation, differentiation and metabolism. Their
number is estimated at about 100 in the human genome (3). These enzymes
function by engulfing in their catalytic site phosphates located on the
tyrosine

CA 02386759 2002-05-17
2
residues of target proteins. The mechanisms underlying the inhibition of
PTPs and the specificity of these peroxo-anionic compounds have been
characterized. The inhibiting potential of PTPs by pVs is a 100 to a 1000
times more powerful than that of oxovanadate (1 ). When compared to known
inhibitors of PTPs such as orthovanadate, molybdate, tungstate and zinc, the
increased inhibiting potential of pV can be explained by the presence of the
peroxide groups, which have the ability to irreversibly oxidize an essential
conserved cysteine residue located in the catalytic domain of practically all
PTPs (4).
The possibility of manipulating the ancillary ligands of pVs is important in
regulating potency and specificity (1 ). The ancillary ligands are more or
less
hydrophilic or hydrophobic and provide the molecule with a specific mode of
action and distribution for the different PTPs. These ligands allow for the
specific targeting of certain PTPs.
B) ANGIOGENESIS:
Almost all tissues and organs develop a vascular network which provides
cells with nutrients and oxygen and enables the elimination of metabolic
waste. Once formed, the vascular network is a stable system with a slow rate
of cellular turn over (5) and yet, the endothelium can become one of the most
rapidly proliferating of all cell types when stimulated (6). Indeed, the
formation of new blood vessels can cause serious physiological
complications. For example, while cornea and cartilage are avascular in
healthy situations, several diseases involving these tissues are complicated
by the massive arrival of blood vessels. Eye angiogenic diseases include
neovascular glaucoma, retrolental fibroplasia, macular degeneration and
neovascularization of corneal grafts. Joint angiogenic diseases include
rheumatoid arthritis and arthrosis. Psoriasis, a chronic condition of the
skin,
also exhibits hypervascularization at the surface of the skin. Finally, solid
tumor growth is critically dependent upon the formation of new blood vessels

CA 02386759 2002-05-17
3
to progress locally and spread all over the body (7,8). The maintenance of
existing blood vessels also requires the regulation of cell replication and/or
differentiation. For example, acute and chronic pathological processes, such
as atherosclerosis, post-angioplastic restenosis and hypertension, involve the
proliferation of different cellular components of mature blood vessels
(endothelial cells, smooth muscle cells, myocytes and fibroblasts).
Several factors, including certain peptides and proteins, can induce a
vascular response in vivo. They are endogenous substances, such as
EGF/TGF-a (Epidermal Growth FactorlTransforming Growth Factor-alpha),
TGF-~3 (Transforming Growth Factor-beta), TNF-a (Tumor Necrosis Factor-
alpha), angiogenin, prostaglandine E2, and monobutyrine (9-16). However,
these factors have almost no mitogenic effect on endothelial cells in culture
(TGF-a, EGF, angiogenine, prostaglandine E2, monobutyrine) and,
paradoxically, inhibit their growth (TGF-~, TNF-a) (10-17). Their angiogenic
action is thus indirect, depending for the most part upon the inflammatory
response (17). Inflammatory cells produce some factors, such as aFGF
(acidic Fibroblast Growth Factor), bFGF (basic Fibroblast Growth Factor),
PDGF (Platelet-Derived Growth Factor), and VEGF (Vascular Endothelial
Growth Factor), which are capable of stimulating the proliferation of
endothelial cells in vitro and angiogenesis in vivo (17-27).
The angiogenic process, as currently understood, can be summarized as
follows: a cell activated by a mutation, lack of oxygen, etc, releases
angiogenic molecules (28-33) that attract inflammatory and endothelial cells
and promote their proliferation. Following the binding of leukocytes to
vascular endothelial cells, the latter reorganize the protein arrangements on
their membranes to activate the angiogenic process (34, 35). During
migration to the target tissue, inflammatory cells release substances that
intensify the angiogenic effect (36, 37). Activated vascular endothelial cells
respond to the angiogenic signal by secreting proteases which digest blood
vessel walls to enable migration toward the target tissue (38-40). Several

CA 02386759 2002-05-17
4
protein fragments produced by the digestion of the blood-vessel walls
intensify the proliferative and migratory activity of the endothelial cells
(41-
43). Finally, the endothelial cells rearrange their adhesive membrane proteins
to generate the formation of capillary tubes.
S
Angiogenesis is thus a complex process consisting of several critical cellular
events (44-46), among which the following may be readily identified:
~ binding of leukocytes to endothelial cells and induction;
~ migration of inflammatory cells to the target tissue;
~ regression of the pericytes of the existing vascular system;
~ dissolution of blood-vessel walls by proteases;
~ endothelial cell migration;
~ endothelial cell proliferation;
~ endothelial cell differentiation and arrangement into a tubular shape;
1 S ~ formation of the capillary network;
~ anastomosis; and
~ initiation of blood flow.
Agents that are known to induce the proliferation of endothelial cells include
sodium orthovanadate (47). The mechanism by which this agent induces an
invasive phenotype in capillary endothelial cells is not clearly understood.
However, the effects of vanadate on cultured cells are similar, in many
respects, to those elicited by PMA (48), bFGF (49), and certain retroviral
transforming proteins, which act by inducing tyrosine-specific protein
phosphorylation (50-53). Consistent with these observations is the finding
that both phorbol ester and various growth factors including bFGF, VEGF
and PDGF stimulate the phosphorylation of cellular proteins on tyrosine
residues whereas vanadate, perhaps by inhibiting tyrosine phosphatase(s),
produces a marked increase in tyrosine phosphorylation.
pVs were expected to promote endothelial cell proliferation, like vanadate.
On the contrary, these agents have been shown to inhibit the proliferation of

CA 02386759 2002-05-17
several cell types (2). In some cases, the cells are arrested at G2/M. The
simplest hypothesis to explain this restriction is that PTPs controlling cell
mitosis are targets for bpV(phen) and pbV (pic) inhibitors. The cdc25 protein
was proposed as one candidate target for pV compounds (2).
5
International Patent Publication WO 95/19177 teaches the use of vanadate
compounds for the treatment of proliferative disorders, metastasis and drug-
resistant tumors. Although these vanadate compounds are stated to be anti-
proliferative and anti-collagenolytic, no indication of any anti-angiogenic
activity has been ascribed to them. This publication further shows that an
anti-tumor effect is observed at dosages of vanadate higher than 5 mM. It is
admitted that a concentration of vanadate compound of 1.3 mM or lower has
no apparent anti-tumor effect.
Montesano et al. (47) teach, on the contrary, that vanadate compounds
cause endothelial cells to proliferate. Hence, their findings would indicate
that
these compounds are pro-angiogenic and not anti-angiogenic.
US Patent 5,716,981 (Hunter et al.) mentions the use of vanadium
compounds, namely oxovanadate, orthovanadate and vanadyl compounds,
in anti-angiogenic applications. However, this reference provides no
experimental evidence to substantiate the anti-angiogenic effects of these
compounds. The compounds are stated to be possibly equivalent to
Paclitaxel (an anti-angiogenic compound described in detail). Since the
remaining body of the prior art suggests that vanadium compounds are pro-
angiogenic, there is no enabling teaching in this patent to validate the use
of
these compounds as anti-angiogenics.
pVs are more powerful anti-tumor molecules than oxo compounds, such as
vanadate. The direct antiproliferative activity of pVs on transformed cells is
known (2). At concentrations found to be ineffective for vanadate

CA 02386759 2002-05-17
6
compounds in International Patent Publication WO 95119177, pV compounds
are efficacious anti-tumor compounds.
Although the anti-tumor activity ascribed to cytotoxicity of vanadium
S compounds is known, their anti-angiogenic activity has not heretofore been
disclosed in relation to either oxo or peroxo derivatives thereof.
C) ENDOTHELINS:
Endothelins (ETs), a family of three isopeptides, acting as important
regulators of the physiological state of mature blood vessels. They are the
most potent vasoconstrictors identified to date. In addition, they are known
to
stimulate the proliferation of endothelial cells, smooth muscles, myocytes and
fibroblasts, as well as the synthesis of various growth factors, including
1S VEGF (54, 55).
ETs are also considered to be angiogenic factors involved in tumor
development (56). Most tumor cells synthesize and secrete ETs (57-61 ).
Patients affected by different cancers show elevated blood concentrations of
ETs (62-64). A reduction in the expression of ET receptor type B (RETg)
decreases the growth of tumor cells incubated in the presence of ETs (64).
As indicated above, ETs promote proliferation and migration of endothelial
cells (65). Expression of ET ligands and of ET receptors (RETA and RETg)
was observed in the endothelial cells of a plurality of tumors (66, 67). ETs
act
2S in an autocrine fashion, promoting local angiogenesis (67). ETs are further
involved in hemodynamic changes that go along with metastatic
development. For example, the ratio of arterial hepatic blood flow to portal
vein blood flow is abnormally high in patients having hepatic metastasis from
colorectal tumors (69). This high blood flow ratio is due to the presence of a
humoral mediator as demonstrated in vivo (70).

CA 02386759 2002-05-17
7
A tumoral vascular bed has no innervation and consequently does not
respond to vasoconstrictive drugs. However, these drugs decrease normal
hepatic blood flow and increase blood flow in tumors. Since ETs are potent
vasoconstrictive and angiogenic factors involved in vascular remodeling and
tumor development, they may be responsible for these altered
hemodynamics. An ET inhibitor may therefore be a valuable tool for
controlling intratumor blood flow and for influencing the growth and
degeneration of tumors.
. D) ANGIOPLASTY:
ETs are also known to stimulate the production of extracellular matrices by
endothelial cells (71, 72). That is frequently observed upon vascular
reconstruction or angioplasty (73, 74). This effect is particularly
deleterious
following vascular trauma due to restenosis. In 30-50% of patients,
restenosis is characterized by the reexpansion of atherosclerotic lesions. The
causes of this vascular disorder are due to a local vascular blockage caused
by cell proliferation, cell migration and extracellular matrix production.
Recently, ETs have been shown to be major players in vascular remodelling,
particularly in such conditions as long term atherosclerosis, cardiac
hypertrophy (congestive heart failure), hypertension (pulmonary and other),
renal problems and certain systemic dysfunctions (75-79). ETs are also
strongly involved in coronary and brain vasospasm leading to ischemia and a
reduced rate of survival (80, 81 ). Thus, because of the potent
vasoconstrictor
and nitogenic activities of ETs, inhibitors of ET production would be expected
to be generally useful as anti-hypertensive and anti-proliferative agents, and
particularly useful prior to, during and after vascular surgery.
E) IMMUNE RESPONSE:
To develop an effective cell-mediated immune response, infected
macrophages (MO) must be able to induce T lymphocyte activation in a

CA 02386759 2002-05-17
8
specific manner. In turn, T cells, by secreting several lymphokines, can
activate M0 for cytocidal functions (82, 84). Several investigators have
demonstrated the importance of cytokines in the control of Leishmania
infection (85-90). Thus, the integrity of M0-T lymphocyte interactions is a
S primordial step for the immune response development, and MQl participation
is crucial to its initiation and support. Tyrosine phosphorylation is a common
event in the initiation of cell proliferation, and its role in signal
transduction
regulating cellular functions of nonproliferative haematopoietic cells is also
well documented (91-97). We have recently demonstrated the importance of
PTPs in NO regulation (98). Correlation between M0 PTP inhibition and an
enhancement of NO production was supported by an increase in M0 PTK
activity and tyrosyl residue hyperphosphorylation (78).
It has been demonstrated that the modulation of protein tyrosyl
1 S phosphorylation states will, in many cases, result in better responses
from
some cells to extracellular stimuli (98, 99-101 ). PTP can play a crucial role
in
the negative regulation of signal transduction culminating in T-lymphocyte
activation. Prior studies have shown that pervanadate (a mixture of vanadate
and hydrogen peroxide) stimulates transcription of the c-fos gene and
accumulation of its mRNA as well as the expression of CD69 antigen and
CD25 (101 ). Also, it was demonstrated that bpV(phen) could induce nucleus
NF-kB translocation in T lymphocyte (102) and increase MO WOS mRNA
expression (98). Rat kupffer cells in culture stimulated with platelet-
activating
factor and vanadate have shown a time- and concentration-dependent
increase in phosphotyrosine in several proteins and of Prostaglandin E2
generation (99). As mentioned earlier, we recently demonstrated that M0
pretreated with bpV(phen) were more responsive to IFNy stimulation as
reflected by the greater amount of NO produced in comparison to Vanadate-
treated and untreated cells (98). Although there is evidence that suggests
that bpV(phen) can be a powerful activator of some immune functions, the
capacity of either oxo or peroxo derivatives to induce cytokines (e.g. IFN(,
IL-
12 and IL-1) and chemokines (e.g. RANTES, MIP-1a,a, MIP-2, IP-10, MCP-

CA 02386759 2002-05-17
9
1 ) recognized for their pivotal role in inflammatory response and the
development of an effective immune response toward a specific antigen, and
thus acting as an adjuvant, has not heretofore been disclosed.
SUMMARY OF THE INVENTION
The present invention provides peroxometallic compounds, such as (pVs),
that are useful against angiogenesis, restenosis and endothelin production,
and as immunomodulators. They have also been found to be suitable for
preventing further growth of established tumors. These molecules comprise
a transition metal (such as vanadium, molybdenum, tungsten) and one oxo or
peroxo groups. Molecules comprising peroxo groups are the more potent
anti-angiogenic molecules. Preferably, the molecules also contain an
ancillary ligand, which includes any molecule capable of binding the
transition
metal atom (usually, through bonds involving oxygen and nitrogen).
Phenanthroline, picolinic acid, bipyridine, oxalic acid, 4,7-dimethyl-
phenanthroline and peptides are examples of such ligands.
All these molecules can be used to inhibit the formation of new blood vessels
and/or control systemic and local levels of endothelins (ETs) in the
reparation
of existing blood vessels.
The molecules containing peroxo anions are more powerful than their oxo
counterparts. Therefore, the former can be used at much lower
concentrations to reduce the toxicity that results from overexposure to
transition metals (2).
Oxo transition metal complexes include oxo complexes such as vanadate,
tungstate, molybdate, and vanadyl complexes, such as the following:
methavanadate (V03-), orthovanadate (V043-), salts thereof, vanadyl
compounds (V02+) like vanadyl acetyl acetonate and vanadyl sulfate.
Similar complexes exist for other transition metals. Other suitable tungsten

CA 02386759 2002-05-17
and molybdenum complexes include hydroxo derivatives derived from
glycerol, tartaric acid and sugars, for example. The peroxo transition metal
complexes include any oxidizing agent capable of combining with the
transition metal. As such, the preferred peroxides comprise the following: t-
5 butylhydroperoxide, benzoyl peroxide, m-chloroperoxibenzoic acid, cumene
hydroperoxide, peracetic acid, hydroperoxiloneic acid, ethyl peroxide,
pyridine peroxide and hydrogen peroxide.
The general structure of the compounds of the present invention is the
10 following:
Y
Z T Z'
L L'
wherein: T is a transition metal selected from the group consisting of
vanadium, molybdenum, tungsten;
Y is oxygen or hydroxyl;
Z and Z' are independently selected from oxygen and peroxide and at least
one of them is peroxide; and
L and L' are any group which can donate an electron pair.
In a preferred embodiment, the transition metal T is vanadium, Y is oxygen, Z
and Z' are peroxide and L and L' are the nitrogen atoms of 1,10-
phenanthroline.

CA 02386759 2002-05-17
11
In another preferred embodiment, the transition metal T is vanadium, Y is
oxygen, Z and Z' are peroxide and L and L' are nitrogen or oxygen atoms of
picolinic acid.
S In yet another preferred embodiment, the transition metal T is vanadium, Y
is
oxygen, Z and Z' are peroxide and L and L' are nitrogen atoms of 2,2'-
bipyridine.
The above pV compounds are potent anti-angiogenics, since they inhibit
endothelial cell proliferation. They further inhibit neovascularization and
the
production of endothelins.
Moreover, administration of the pV compound bpV(bipy) in the rat model
revealed a significant reduction in the degree of post-angioplastic vascular
remodeling of the carotid artery. Additionally, the pV compound bpV(phen)
was found to be a potent immunomodulator based on its capacity to induce
cytokine and chemokine gene expression, to enhance cellular recruitment in
response to an agonist and consequently act as an adjuvant in the context of
vaccination.
In one specific embodiment, the present invention relates to the inhibiting
action on tumor growth of bpV(phen), demonstrating efficiency in vivo. In
addition it is shown that bpV(phen) has also the capacity to inhibit the
migration of tumor cells in vitro.
Other objects, advantages and features of the present invention will become
apparent upon reading the following non-restrictive description of preferred
embodiments thereof, with references to the accompanying drawings.

CA 02386759 2002-05-17
12
DESCRIPTION OF THE PREFERRED EMBODIMENTS OF THE PRESENT
INVENTION
This invention will now be described by referring to specific embodiments and
the appended Figures, which purpose is to illustrate the invention rather than
to limit its scope.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1: Peroxovanadium compounds inhibit the proliferation of endothelial
cells. Human umbilical vein endothelial cells (HUVECs) were extracted with
collagenase-controlled digestion. Pure HUVECs were used before the fourth
passage (trypsin-EDTA at each passage). The cells were analyzed for their
capacity to incorporate di-acetyl LDL and to be labelled with factor VIII-
related antigen. Endothelial cells were plated at a density of 2500 cells/cm2
in a sterile plate coated with gelatin. Cells were cultured in complete medium
(M199: heparin (90mg/ml) or L-glutamine (2mM), bicarbonate, FBS (20%)
and ECGS (100mg/ml) for 24 hours to ensure cell adhesion. Then, cells
were washed 3 times with PBS and culture medium was added according to
experimental conditions. The last PBS wash was considered as time t=0..
Cell proliferation was evaluated with the amount of DNA present in the petri
dishes. Each experiment was performed in triplicate. The culture medium
was changed daily. After 96 hours in culture, cells were lysed with Na-
Citrate-SDS solution and incubated with Hoescht 33358. Samples were read
at 365 nm with a spectrofluorometer. The results show a dose-response
inhibition of endothelial cell proliferation with the pV compounds. The
approximate IC5o is 2mM for bpV(phen) and 3.5 mM for bpV(pic).
Figure 2: A and B show an anti-angiogenic response to bpV(phen),
orthovanadate (Van) and protamine (Prot) using a vascularization test on the
vitelline membrane of chick embryos. Each point represents a minimum of 15
embryos (15 to 28). In Figure 2A), neovascularization was assessed using
N/+ scoring system; the proportion of embryos showing anti-angiogenesis

CA 02386759 2002-05-17
13
increased with dosage, bpV(phen) being the most potent agent. In Figure 2B,
neovascularization was assessed using the 1-2-3 scoring system; the
angiogenic score decreased with dosage, bpV(phen) being the most potent
agent.
Figure 3: Micrographs of the cell migration from aortic rings embedded in
fibrin (x12). Compared to the sample control (A), cell migration was impaired
in the presence of doses of bpV(phen) (1, 3.5 NM; B, C) with a strong
inhibition at 3.5 NM (C). In the presence of bpV(pic) (3.5, 10, 25 NM ; D, E,
F),
inhibition was seen at 10-25NM. In the presence of orthovanadate, partial
inhibition was reached at 25 NM (G) with no effect at 3.5 NM (H). The
migration of well-defined microvessels in the presence of bpV(pic) is evident
at 10 NM, while in the control sample very thin cell strands were frequently
observed.
Figure 4: Quantification of the cell migration from aortic rings in the
presence
of orthovanadate (plain bars); bpV(phen) (light gray bars); and bpV(pic) (dark
gray bars). The control appears as an empty bar. Means and standard errors
of the means are presented. The Student-Newman-Keuls method was used
for statistical analysis (p value=0.001 )
Figure 5: bpV(phen) inhibits angiogenesis in vivo in the s.c. Matrigel assay.
Graph showing the results of an experiment expressed as the number of
cells that migrated in three microscopic fields for each s.c. Matrigel plug.
Fields were equidistant from the edge of the plug. (1 ) Four mice that did not
receive any treatment after the Matrigel plug implantation. (2) Four mice that
received daily administration of 10 Ng bpV(phen) during the 7 days post-
implantation. (3) Four mice that received daily administration of 50 Ng
bpV(phen) during the 7 days post-implantation. (4) Four mice that received
daily administration of 100 Ng bpV(phen) during the 7 days post-implantation.
(5) Four mice that received daily administration of PBS during the 7 days
post-implantation.

CA 02386759 2002-05-17
14
Figure 6: Neutrophils, eosinophils and MQ~ recruited in air pouch exudates of
bpV(phen)-treated BALB/c mice in response to L. major promastigotes intra-
pouch injection. Air pouch exudates were collected from animals injected
with endotoxin-free PBS, LPS (20 ~g/ml), L. major and bpV(phen) (500 nM
S i.p., 2 hour) + L. major, 6 hr post-inoculation. Each time point represents
the
mean ~ SD (n= 4 mice) of 2 experiments similarly performed. Differences
observed for specific leukocytes population recruited were significant (p <
0.01, Student's t test) over their respective control.
Figure 7: In vivo Leishmania-induced pro-inflammatory mediator generations
in pV-treated mice. (A) Nitrate level determination in air pouch exudates
collected 6 hr post-inoculation of endotoxin-free PBS, L. major and
bpV(phen) followed by L. major has been performed. Values are the mean ~
SEM of 2 experiments performed independently. (B) Evaluation of pro-
inflammatory cytokines (IL-6 and IL-1(3) and chemokines (MCP-1 and MIP-2)
secreted in air pouch exudates collected as described above were measured
by ELISA as described in more detail below. Values are the mean ~ SEM of
2 experiments performed independently. Levels of pro-inflammatory
molecules measured in exudate supernatants were significantly augmented
(p < 0.01, Student's t test) in response to L. major infection and
significantly
further increased by bpV(phen) treatment over their respective control. IL-6,
IL-1[3, MCP-1 and MIP-2 secretion were respectively 12.8-; 2-, 8- and 6.8-
time more secreted in L. major -injected bpV(phen)-treated animals
compared to L. major alone. PBS control was equal to background.
Figure 8: In vivo bpV(phen)-induced cytokine gene expression in spleen of
BALB/c mice. Expression of Th1 (IFN-(, IL-2, IL-12) and Th2 (IL-4 and IL-10)
cytokines were monitored in spleen of BALB/c mice in response to bpV(phen)
over a 24-hr period. Expression of cytokine genes has been monitored using
a multi-probe RNase protection assay as described in more detail below.
Results obtained are representative of 3 experiments performed
independently.

CA 02386759 2002-05-17
Figure 9: Effect of bpV(phen) on chemokine mRNA expression in murine
B10R macrophages. (A) Cells were incubated for 2 h with or without various
concentrations of bpV(phen) (1-50 pM). Total RNA was isolated and
analyzed by RNase protection. (B) Kinetic expression of chemokine genes in
5 murine B10R macrophages. Cells were incubated for increasing period of
time (1-4 h) with or without bpV(phen) at 10 NM. Following extraction of total
RNA, RNase protection assay was performed. Free RNA probe is shown in
the far left lane. The fold increase was calculated from the densitometry of
the autoradiogam.
Figure 10: Use of bpV(phen) as an adjuvant in Leishmania vaccination trial.
BALB/c mice were inoculated with soluble Leishmania major antigen (SLA;
100 Ng) with or without daily injection of bpV(phen) (500 NM) for 5 days. Two
weeks later animals received a second set of treatment as above. Control
animals received either PBS or bpV(phen) alone without any SLA. At 4
weeks post-vaccination, animals were challenged with 5 million Leishmania
major stationary phase promastigotes injected in the right hind footpad.
Progression of infection was followed by measuring footpad thickness with a
caliper on a weekly basis. Differences measured for reduced footpad
thickness were significant (p < 0.01, Student's f test, n=5-10 animals per
group) over their respective controls.
Figure 11: Illustration of the antitumor activity of bpV(phen) in vitro using
PC3 prostate cancer cells. Collagen gels containing the PC3 cells were
prepared according to the method of Esdale and Bard (1972). Briefly, stock
collagen solution (3.5mg/ml in acetic acid 0.02N; Rat tail) was added to a
mixture composed of culture medium (5x), fetal bovine serum (FBS),
bicarbonate (0.26M), and was neutralized with 0.1 N NaOH. A cell suspension
(1.42 x 106 cells/ml) was mixed into the collagen-medium mixture to obtain a
final concentration of 2.5x105 cells/ml.

CA 02386759 2002-05-17
16
A DMEM medium, having a normal concentration of glucose and without
phenol red, was used. After gelification (within 1 h) of the collagen mixture
containing the cells, the gels were removed from their culture wells (mould)
and interwoven into a receptor hole prepared in a fibrin gel, as previously
described (103). The fibrin gel was made from a 0.3 % fibrinogen solution in
Hank's balanced salt solution. Fibrin was allowed to polymerize with thrombin
(stock solution at 1.75mg/ml) at a ratio of 1:003 v/v fibrin to thrombin. The
collagen-fibrin complexes were then covered with serum-supplemented
medium according to the cell types. An inhibitor of plasminogen activator
(Trasylol, Parke Davies) was added into the medium at 10 pl/ml (100UIml).
Cell behavior was periodically monitored over 15 days of culture.
In this model, collagen is believed to mimic the tumor stroma, and fibrin is
well recognized as the primary matrix for cancer cell expansion and migration
(104). bpV(phen) was used at 2, 5 and 10 ~M; the compound was renewed
daily over a 2 week period. Phase contrast microscopic observations and
micrographs were taken after 15 days, and samples were prepared for
histological sections. Histological sections were stained with periodic acid
Shiff stain to enhance the matrix contrast.
Figure 12: Illustration of the antitumor activity of bpV(phen) in vitro using
ZR-
75 breast cancer cells. Experiments were done as described for the figure
11 except that DMEM medium having a high glucose concentration and
without phenol red was used. 10% FBS was used instead of 5% FBS. The
media was supplemented with 109 M estradiol (final concentration).
Figure 13: Figures 13A and 13B illustrates the antitumor activity of
bpV(phen) in vivo.
Figure 14: In vitro antitumor activity of lymphocytes pretreated with
bpV(phen). PC-3 cancer cells embedded in a collagen gel, grew as a
"primary tumor". Some cells migrated from the primary tumor toward the

CA 02386759 2002-05-17
17
fibrin gel, forming front edges that can be quantified. Small clumps of cells
progressively appear in the fibrin gel and we assume that these cell
extension are representative of the invasive potential of the cancer cells. In
the control panel (left), PC-3 cells migrated slightly from the primary tumor
and formed extensive secondary tumors in the fibrin gel. In the presence of
bpV(phen)-treated lymphocytes no secondary tumor was observed and there
was no migration front. In addition, the primary tumors appeared less dense
than in the control.
Definitions
In order to provide a clear and consistent understanding of terms used in the
present description, a number of definitions are provided hereinbelow.
Unless defined otherwise, the scientific and technological terms and
nomenclature used herein have the same meaning as commonly understood
by a person of ordinary skill to which this invention pertains.
For the purposes of the present application, the term "animal" is meant to
signify human beings, primates, domestic animals (such as horses, cows,
pigs, goats, sheep, cats, dogs, guinea pigs, mice, birds, fish etc.).
From the specification and appended claims, the term "therapeutic agent"
should be taken in a broad sense so as to also include a combination of at
least two such therapeutic agents.
For administration to humans, the prescribing medical professional will
ultimately determine the appropriate form and dosage for a given patient, and
this can be expected to vary according to the chosen therapeutic regimen,
the response and condition of the patient as well as the severity of the
disease.

CA 02386759 2002-05-17
18
Compositions within the scope of the present invention should contain the
active agent (e.g. compound) in an amount effective to achieve the desired
therapeutic effect while avoiding adverse side effects. Typically, the
compounds of the present invention can be administered to mammals (e.g.
S humans) in doses ranging from 0.001 to 50 mg per kg of body weight per day
of the mammal which is treated. Pharmaceutically acceptable preparations
and salts of the active agent are within the scope of the present invention
and
are well known in the art (Remington's Pharmaceutical Science, 16th Ed.,
Mack Ed.). The dosage will be adapted by the clinician in accordance with
conventional factors such as the extent of the disease and different
parameters from the patient. Typically, 0.001 to 50 mg/kg/day will be
administered to the mammal.
A) PEROXOVANADIUM COMPOUNDS and ANGIOGENESIS
1. Peroxovanadium compounds inhibit the proliferation of endothelial
cells and the formation of tubular structures
Human umbilical vein endothelial cells (HUVECs) were extracted with
collagenase-controlled digestion, as previously described (105). Pure
HUVECs were used before the fourth passage (trypsin-EDTA at each
passage). The cells were analyzed for their capacity to incorporate di-acetyl
LDL and to be labelled with factor VIII-related antigen.
Endothelial cells were plated at a density of 2500 cells/cm2 in a sterile
plate
coated with gelatin. Cells were cultured in complete medium (M199: heparin
(90 ~g/ml) or L-glutamine (2mM), bicarbonate, FBS (20%) and ECGS
(100 ~g/ml)) for 24 hours to ensure cell adhesion. Then, cells were washed 3
times with PBS and culture medium was added according to experimental
conditions. The last PBS wash was considered as time t=0.
Cell proliferation was evaluated with the amount of DNA present in the petri
dishes. Each experiment was performed in triplicate. The culture medium

CA 02386759 2002-05-17
19
was changed daily. After 96 hours in culture, cells were lysed with Na-Citrate-

SDS solution and incubated with Hoescht 33358. Samples were read at 365
nm with a spectrofluorometer.
S The results show a dose-response inhibition of endothelial cell
proliferation
with the pV compounds (Figure 1 ). The approximate IC5o is 2 ~M for
bpV(phen) and 3.5 pM for bpV(pic).
Alternatively, HUVECs cultured in a fibrin matrix can form 3-dimensional
tubular-like structures in the presence of serum (103). This assay was
performed in the presence of either bpV(phen), bpV(pic) or vanadate to
assess the influence of each one of these compounds on HUVEC
differentiation and organization (Table 1 ). HUVECs were seeded on the
bottom of gelatin-coated wells at high density to provide a confluent
monolayer at 48 hours.
Then, 50 000 HUVECs/ml were embedded in fibrinogen solution prior to
polymerization. The fibrin matrix was covered with culture medium containing
the molecule to be tested, while culture medium without any of these
molecules served as controls. Cell behavior was observed periodically by
phase contrast microscopy. After 21 days in culture in the presence of 1 ~M
bpV(phen), cord-like structures (or cords), tube-like structures (or tubes)
and
stellate structures (or Stell.struct.) were observed. At higher doses (2 and
3.5
uM), fragmented cord-like structures were apparent. In the presence of
bpV(pic), cords and tubes were observed at 1, 2 and 3.5 pM. In the presence
of orthovanadate, cords were still apparent at 10 ~M, and at 25 pM dead
cells were sparsely distributed. These results suggest that pV compounds
interfere with endothelial cell organization and terminal differentiation.
Furthermore, the nature of the ancillary ligand is important since bpV(phen)
is
more potent in inhibiting tube formation than bpV(pic).

CA 02386759 2002-05-17
The results show that vanadate has an anti-angiogenic effect inasmuch as
the stellate structures are affected (organization and terminal
differentiation).
Vanadate is at least 3 and 5 times less potent than bpV(pic) and bpV(phen),
respectively. The anti-angiogenic effect observed with vanadate is in
5 contradiction to the teachings of Montesano (47).
Indeed, it was observed that at low doses, all the tested vanadium
compounds seemed to be pro-angiogenic, while at higher doses (i.e., at
almost double the dose), the same compounds were clearly anti-angiogenic.
10 The type of radical L, L' greatly affects the potency of the vanadium
compounds; for example, phenanthroline was found to be twice as potent as
picolinic acid.

CA 02386759 2002-05-17
21
TABLE I: Effects of bpV(phen)i, bpVi(pic) and Vanadate on HUVEC
Differentiation and Orgianization
Doses Stell.
(uM) cords tubes struct.


bpV 0 +++ +++ +++
(phen)


+++ +++ +++


++ + _


3.5 - - -


bpV (pic)0 +++ +++ +++


+++ +++ +++


2 +++ +++ +++


3.5 ++ + _


van 0 +++ +++ +++


2.5 +++ +++ +++


10 +++ +++


25 t 1'


t Dead cells

CA 02386759 2002-05-17
22
2- Rat aortic ringassay
Rat aorta rings were embedded in fibrin matrix as previously described (107).
Thoracic aortas excised from adult rats were rinsed in Hank's balanced salt
solution (HBSS), and cleansed of periadventitial. One-mm long aortic rings
were sectioned and rinsed in culture medium. Each of the aortic rings were
then embedded in fibrin gel matrix. Migration of microvessels in fibrin gel
was
quantified by measuring the distance from the external surface of rat aortic
rings towards the migrating vessels. Measurements were performed at day
by digitizing morphometry with a NIH image analysis system (Fig. 3 and
10 4).
3- Peroxovanadium compounds inhibit neovascularization ex ovo
a. Definition of the test-s ski tem:
The normal development of a chick embryo involves the formation of an
external vascular system which is located in the vitelline membrane and
15 which carries nutrients from the vitellus (yolk of the egg) to the
developing
embryo. When placed onto the vitelline membrane, anti-angiogenic
substances can inhibit blood vessel development in the vitelline membrane.
To facilitate access to the vitelline membrane, chick embryos were
transferred to a sterile culture box (Petri dish) and placed in a humidity-
and
temperature-controlled incubator. Embryos could then develop in this ex ovo
condition for several days.
An aliquot of the tested compound was mixed with a methylcellulose solution
and allowed to air-dry into thin discs. Methylcellulose forms an inert matrix
from which the tested compound can diffuse slowly. Methylcellulose discs
containing the tested compound were placed on the external border of the
vascular perimeter of the vitelline membrane where the angiogenic process
was still active.
The effects of the discs containing the tested compound on proximal vascular
development were, in all cases, compared with those of discs containing
control buffer. The discs were placed on the embryos' vitelline membrane on

CA 02386759 2002-05-17
23
Day 0 or Day 1 of the ex ovo growth process; at this point, only beginnings of
the main blood vessels are invading the vitellus. The embryos were then put
in culture conditions until vascularization was assessed (approximately 24 h).
Control buffer- and compound-containing discs were in all cases added
simultaneously on the vitelline membrane of the same embryo. Both discs
were arranged in a symmetrical fashion with respect to the cephalo-caudal
axis of the embryo in order to minimize inter-individual variations when
comparing the tested compounds with controls.
b. Anti-angriogienic activity:
Embryonic vascularization tests (EVTs) were performed using different
concentrations of protamine (5 to 20 fig) as a positive control or a tested
compound (0.001 to 10 fig). After one day in culture, the level of
vascularization in the area covered by the discs was graded by a pair of
scientists in the usual blind fashion. To facilitate the location of the
discs,
black O-rings were placed around them just after their placement on the
vitelline membrane. The evaluation scale for the EVTs was based on two
different scoring systems.
Assessment of blood vessel formation:
Blood vessel formation was assessed in a blind fashion. The areas of the
vitelline membranes that lay beneath the methylcellulose discs were scored
for the degree of vascularization, using two scoring systems ("N/+" or " 1-2-
3"). The following selection criteria were used:
NI+ system:
A score of "N" for "Normal" was attributed when all the following conditions
were met:
Blood vessels in the evaluated area grew along their path with no abnormal
deviation. Collateral branching density was normal and the growth path of the
lateral branches was also normal.

CA 02386759 2002-05-17
24
A score of "+" was attributed when at least one of the following conditions
was met:
~ Major blood vessels grew across the evaluated area but their paths were
clearly affected (winding).
~ Major blood vessels grew across the evaluated area but collateral
branching density was clearly diminished.
~ Major blood vessels penetrated the evaluated area but their growth path
rapidly deviated.
~ A kink was observed in the blood vessel.
~ Major blood vessels penetrated the evaluated area but were stunted. No
growth was observed beyond that point.
~ A drastic deviation in the growth path occurred when major blood vessels
reached the proximal ridge of the disc.
1-2-3 grading scale system:
A score of 3 was attributed when the following conditions were met:
~ Blood vessels, present in the evaluated area, grew along their paths with
no abnormal deviation. Collateral branching density was normal and the
growth paths of the lateral branches were also normal.
A score of 2 was attributed when at least one of the following conditions was
met:
~ Major blood vessels grew across the evaluated area but their paths were
clearly affected (winding).
~ Major blood vessels grew across the evaluated area but collateral
branching density was clearly diminished.
A score of 1 was attributed when at least one of the following conditions was
met:

CA 02386759 2002-05-17
~ Major blood vessels penetrated the evaluated area but their growth paths
were rapidly deviated.
~ A kink was observed in the blood vessels.
~ Major blood vessels penetrated the evaluated area but were stunted. No
5 growth was observed beyond that point.
~ A drastic deviation in the growth paths occurred when major blood
vessels reached the proximal ridge of the disc.
A score of 3 meant that blood vessel development was normal whereas a
10 score of 1 indicated the greatest degree of angiostatic activity.

CA 02386759 2002-05-17
O
_N
N
M r- t~ 00 Cfl d'


M N N ~ - N N
C


O V


~ r- f' O tf~
R o Cfl ~t


r- tt~ t~- cD - N


c


E tn N N ~' N Op CO
~
C


c~ Q Z N T- ~ t- N N
Q
cC


O . n


O


a r.. M ~ oo o


M N N


_.


V o ~ O 1' c
O


t0 ~ O tf~ 00 (D ~ i
~


v N
~
tQ


to N
C


c r- N N N U
~


a Z ~- ~ r- r- ~ ~ c
c~


N
O O


.~ lI~ .


. vi
M ~ ~ r- ~ ~ ~' C


~ c6


. C
:
:


, O
.
O


d .a M o ~ i I~- i
~


QE ea '
,
o


N '


> Y c ~, c
~


Q Z . . ~- . . . _
ca 3



0
m W n


O ~ M ~ ~ ~ ~ N N



O ~


O
C7 ~ ~ O O Q7


Q a nt o ~ ~ a0 ~ N r- N
= ~ .N
~


o
c


p '+


r
1~3~ N ~ ~ M
tIlCV


00
a Z i ~ ~ ~ ~ e-
nT __.



d M O f~ M O


M N ~ N ~ N N



N O ~ C


O o M cp N r-


\ N ~ d~ ' '- M


O _ ~'
r


C vi


O O ~ ~
~ ~u~


O ~
O


fn ~ N M M
c


Q Z e- . r' ' ~ r-
Q


~
- O
.



O C C C C


V ~ _ _ _ _ ~ ~ '~
O


R ~. ~ ~ ~ ~ L t r:


r. ;~ aT ~_ ,
N


O O O O
a ' ' ' '


i- n a a a o
- z a~ . c _ c
~ ~


c
c


a


11


0


z
'-v,


c




CA 02386759 2002-05-17
O N ~ d" ~' O O


N T N N N ~ N


tfj ~ t' N N CEO T


00 c~ ~ ~ CO N
N N ~ T - N N tt~


O ~ O


e- , 1 , T N


O r-


, O , , , lf) a0


O
, T 1 1 1


00 ~ d' O


, N N N N N


~f o0 O
t' N N f~ O


tn ~ c~ ~ O


1 , ~- .-- ~-- T
-


N


N T O


CV r- , . , 1 N


O


tf~ O


T . 1 1 I r..


N O


r- ~ , , , 1 O


a0 Cfl


c- r- , , 1 1


r- CD


CD O , , , 1


T


T T 1 1 1 1


T T


1 n O T


C C O O T



r



r
.n o ~ 0 0 o O U



CA 02386759 2002-05-17
28
A dose-response inhibition was obtained with protamine, bpV(phen), and
orthovanadate (Figure 2). The approximate IC5o is 0.1 pg for bpV(phen),
0.6 ~g for orthovanadate, and 11 ~g for protamine (regardless of the scoring
system). A summary of the experimental data collected is shown in Table II.
The results show that bpV(phen) is a potent inhibitor of the angiogenic
process. Prior art on the subject (vanadate) taught the opposite, namely that
vanadium compounds promote angiogenesis.
4- In vivo Matrigel slug assay
This assay was performed as previously described (107). Briefly, Matrigel
(liquid at 4°C) was mixed with 200 ng/ml ECGS and injected s.c. into
four
C57B1/6N female mice (1 ml/mice). After injection, the Matrigel polymerized
to form a plug. After 7 days, the animals were sacrified, and the Matrigel
plugs were removed and fixed in 10% neutral-buffered formalin solution
(Sigma Chemical Co.) and embedded in paraffin. Histological sections were
stained with Masson's trichrome and angiogenesis was scored by counting
the number of cells that migrated in three microscopic fields for each s.c.
Matrigel plug.
Experiments were performed to determine whether bpV(phen) inhibits vessel
formation in vivo. Animals were divided into five groups (4 mice/group): two
control groups that received no treatment or daily i.p. PBS injection,
respectively, and three groups that received daily administration of various
doses (10, 50 and 100 pg) of bpV(phen), from day 1 to day 7 (intraperitoneal
daily administration of 100 NI solution). The results show that bpV(phen) is a
potent angiogenesis inhibitor in vivo (Fig. 5). Daily administration of 10
pg/mice or higher doses decreased by half the number of endothelial cells
having invaded Matrigel plugs.

CA 02386759 2002-05-17
29
The angiogenic potential of pV compounds was tested using in vifro, ex ovo,
and in vitro systems of analysis. The results show that in contrast to what
might have been expected from the teaching of the prior art, pV compounds
are potent inhibitors of the angiogenic process (see below), and this includes
high doses of vanadate.
B) PEROXOVANADIUM COMPOUNDS and THE PRODUCTION OF
ENDOTHELINS
The data presented in Table III show the effect of bpV(phen) on the plasmatic
levels of ETs. In this study, rats were injected intraperitoneally with
bpV(phen) (0.5 mg/100g b.w.) 16 hours before the administration of either
insulin or vehicle. Two minutes following insulin administration (1.5 ~g/100g
b.w, or vehicle), the plasma levels of ETs were determined. Insulin
administration induced a strong increase of seric ET concentration (54). This
increase was completely abolished by bpV(phen). In addition, bpV(phen)
decreased the insulin-stimulated levels of plasmatic ETs below control levels.
These results suggest that bpV(phen) inhibits the insulin-induced release of
ETs that subsequently can lead to diabetic complications like vasculopathy
and nephropathy.
TABLE 111: Effect of bpV i(phen) on Endothelin Plasma Levels
Plasma endothelins
(pg/ml)



Control 113.41 10.91



Insulin 253.10



Insulin, bpV(phen) 77.68 3.08


ETs (ET-1, ET-2, ET-3) were measured by RIA (Amersham kit RPA 555)
after lyophilization and extraction on C2 columns (500 mg). Results are mean
~SEM, p<0.0209, control (n=4) vs Insulin, pV (n=3).

CA 02386759 2002-05-17
C) PEROXOVANADIUM COMPOUNDS and RESTENOSIS
The data presented in Table IV show the effect of the pV compound
bpV(bipy) on restenosis. Male Sprague-Dawley rats (325-350g) were
5 anaesthetized with Rogarsetic, a mixture of ketamine hydrochloride and
xylazine hydrochloride. The animals were divided into two groups of 14-31
rats that underwent balloon angioplasty of the left carotid artery, with or
without treatment (108). The treated group received bpV (bipy) (10 mg I kg I
i.p. b.i.d.) for 14 days, starting on the day of the angioplasty. Every animal
in
10 each group underwent balloon angioplasty of the left external carotid
whereas the contra-lateral carotid served as a control for each animal. The
left carotid artery balloon angioplasty was performed under aseptic
conditions. The left external carotid was isolated and an arteriotomy was
performed. A 2F Fogarty arterial embolectomy catheter was then inserted in
15 the left common carotid and positioned near the aortic arch. The balloon
was
then inflated and retracted with a twisting motion to its insertion point.
This
procedure was repeated twice and the external carotid was ligated. There
were no differences in the lumina (L), intima (I), media (M), the ratio of I/M
and calculated value of (M - L / P) as a measure of neointimal proliferation
20 (NIP) between normal, non-ballooned carotid artery of treated and non-
treated rats, serving as controls (data not shown). Treatment caused a 32%
decreased in NIP and the I/M ratio, a 21 % reduction in the thickness of the I
and a 28% increase in the opening of the L. Thus, treatment with bpV (bipy)
revealed a significant reduction in the degree of post-angioplastic vascular
25 remodeling of the carotid artery in the rat model.
The above results illustrate the antiangiogenic and anti-restenosis potential
of
vanadium compounds. The administration of pV compounds could be used to
control the progression of several angiogeno-dependent conditions as well as
30 post-angioplastic vascular remodeling. Since vanadates are anti-tumor
compounds, the present pV compounds should function as anti-tumor
therapeutic agents, and part of this activity should be due to an anti-

CA 02386759 2002-05-17
31
angiogenic effect. Pharmaceutical compositions would comprise potent pV
compounds capable of achieving an extracellular concentration of about 0.1
to 100 ~M, preferably 2-40 ~M. In cats, a dose of 20 moles per kilogram
was successful in reversing the endothelin increase after insulin
administration. Moreover, pV compounds, which as previously stated are
more potent and provoke less side effects than the vanadium "oxo"
compounds, can be administered to achieve the desired dosage. For
example, see in vivo effects on blood glucose levels have been reported (1,
3).
TABLE IV: Effect of bpV (bipY) on Restenosis
NIP I M I / L
mm mm2 M mm2


Control 0.064 0.138 0.090 1.546 0.090


bpV(bipy)0.045 0.112 0.104 1.086 0.112


p < x 0.002 0.04 0.005 0.003 0.003


Abbreviations: I, intima; M, media; NIP, neointimal proliferation; L, lumina.
D) PEROXOVANADIUM COMPOUNDS and THE IMMUNE RESPONSE
The present invention provides pV compounds as novel immunomodulator
and adjuvant molecules. The pV compound bpV(phen) can be used as an
immunomodulator in vitro (macrophage; chemokine genes expression) and in
vivo (murine model; cytokine genes expression, cytokine and chemokine
generations and cellular recruitment in response to various stimulation), and
as an adjuvant to maximize the protective effect of known Leishmania
antigen based vaccine. The above bpV(phen) compound is a potent
immunomodulator based on its capacity to induce cytokine (e.g. IL-12, IFN(,
IL-1~3) and chemokine (e.g. RANTES, MIP-1a,(3, MIP-2, IP-10, MCP-1) gene

CA 02386759 2002-05-17
32
expression to enhance cellular recruitment in response to an agonist and
consequently act as an adjuvant in the context of vaccination.
1-bpVl~phen) treatment enhance pro-inflammatory molecules secretion
S and polymorphonuclear recruitment in response to Leishmania
infection
With the use of a murine air pouch model, we investigated early inflammatory
events that occur following L. major skin injection in bpV(phen)-treated
animals (Figure 6). As shown in Figure 6A, both L. major and LPS (positive
control) could induce significant leukocyte recruitment in air pouch exudates
as measured 6 hours post-injection. Of interest, bpV(phen)-treated BALB/c
mice, in response to L. major promastigotes (107 parasites/ml) intra-pouch
injection, manifested a five times greater recruitment of cells within the
same
time period. In addition, differential counts performed on Diff-Quick-stained
cytospin preparations from these cells (Fig. 6B) revealed that --70% of
recruited leukocytes were neutrophils (18% eosinophils and 12%
macrophages), whereas in untreated animals receiving L. major
promastigotes ~48% of recruited cells were neutrophils with the rest of the
cell population consisting of eosinophils (26%) and macrophages (26%). This
data revealed that bpV(phen) is a good modulator of the early inflammatory
response and could have a major impact on the progression of L. major
infection since the dramatic increase in inflammatory cell recruitment
consisted mainly of neutrophils, already recognized for their importance to
restrain Leishmania pathogenesis (109).
In parallel to this experiment, in vivo generation of Leishmania-induced NO
and pro-inflammatory mediators in bpV(phen)-treated mice were measured in
air pouch exudates (Figure 7).
The present set of data reinforce the idea that bpV(phen) is an excellent NO
modulator as revealed by its enhanced generation in response to L. major
infection (Fig. 7A). This elevated NO generation not only further increased

CA 02386759 2002-05-17
33
the microbicidal activity observed in bpV(phen)-treated animals, but also
partly explained the strong inflammatory response observed in bpV(phen)-
treated animals since NO has been recognized as an important mediator of
inflammation and regulator of neutrophil migration (110, 111 ). In addition,
bpV(phen)-treatment has shown elevation of some pro-inflammatory
cytokines (IL-6 and IL-1~i) and chemokines (MCP-1 and MIP-2) measured in
air pouch exudates of animals inoculated with L. major promastigotes (Fig.
7B). MIP-2 chemokine is recognized as a specific chemoattractant for
neutrophils (112) whereas MCP-1 has been shown to be a powerful
monocyte/macrophage recruiter to sites of inflammation. Additionally, the
pro-inflammatory cytokines IL-1(3 and IL-6 are well known chemokine
modulators playing a pivotal role in the development of inflammation (113)
including the regulation of MCP-1 and MIP-2 expression (114).
1S 2- Up-regulation of IL-12, IL-2 and IFN~~ mRNA expression in spleen of
bpV(phenJl-treated animals
As shown in Figure 8, in vivo bpV(phen)-induced cytokine gene expression-
monitored with the use of a multi-probe RNAse protection assay-in spleen of
naive mice was significantly up-regulated for up to 24 hr (i.e. IL-12, IL-10,
IL-
2 and IFN() in comparison to animals injected with PBS. Our observation
provides evidence that the PTP inhibitor bpV(phen) is a powerful
immunomodulator favoring the activation of protective Th1 type cytokines
recognized to modulate NO generation in vivo resulting in control over
cutaneous leishmaniasis (115, 116). Additionally, whereas little is known
concerning the role of PTPs on the regulation of cytokines, the present
observation revealed that different signaling events being or not under the
control of PTPs conduct to cytokine genes regulation. Overall, these last
experiments clearly demonstrated the capacity of bpV(phen) to up-regulate
several protective inflammatory and immunological functions of the host in
response to a pathogen.

CA 02386759 2002-05-17
34
3- Effect of bpV(phen) on chemokine gene expression
Various concentrations of bpV(phen) were added to B10R macrophages for 4
h. mRNA expression was measured by RNase protection assays. RANTES,
S MIP-1a/~i MIP-2, IP-10 and MCP-1 chemokine mRNA levels increased in
response to bpV(phen) (Fig. 9A). MCP-1, MIP-2, MIP-1~3 and RANTES were
induced by the addition of bpV(phen) in a dose-dependent manner whereas
IP-10 and MIP-1a were expressed at markedly lower concentrations (10 pM).
Thus, bpV(phen) used at 10 ~M seems to be the most effective dose for the
induction of chemokine gene expression. In addition, we noted that their
expression was were differently modulated by bpV(phen) treatment,
suggesting that different type of PTPs must play a specific role in the
regulation of various chemokines.
To determine the kinetics of chemokine expression following pV compound
treatment, B10R macrophages were incubated with 10 DM bpV(phen) for
increasing time intervals. Total RNA was extracted from macrophages at
various time-points and subjected to RNase protection assay. There was a
transient induction of MIP-1a, MIP-2, IP-10, and MCP-1 chemokines mRNA,
reaching maximum levels at 4 h and rapidly declining afterward (Fig. 9B),
whereas for MIP-1 ~3, the optimal expression was achieved at 8h post
treatment. However, the expression of RANTES mRNA increased in a time
dependent manner over 24 h (Fig. 9B). Thus, conditions for the induction of
chemokine expression in response to bpV(phen) seems to vary with the
different chemokines.
These results clearly showed that bpV(phen) had a stronger effect on some
chemokines than on others. Evaluation of mRNA expression by densitometry
analysis (data not shown) demonstrated a greater induction of MIP-2>MIP-
1a>MCP-1 in presence of bpV(phen) in comparison to MIP-1~3 >RANTES>IP-
10.

CA 02386759 2002-05-17
We report that modulation of host PTPs by bpV(phen) was effective in
inducing RANTES, MIP-1a, MIP-1~i MIP-2, IP-10 and MCP-1 chemokine
gene expression in B10R murine macrophages. However, its action varied
according to each chemokine, demonstrating a more potent effect on specific
5 chemokines. Indeed, RNase protection assays have shown a greater
induction of MIP-2> MIP-1a> MCP-1 by adding bpV(phen) compared to MIP-
1 ~i >RANTES>IP-10. Thus, our results designate PTPs as important negative
regulators of the signaling process implicated in the chemokine production
when they are activated. This is consistent with several studies where PTPs
10 negatively regulate many cellular signaling such as B-cell receptor (BCR)
(117) and erythropoietin receptor (118).
4- Use of bpVi(phen) as adjuvant in the context of Leishmania vaccine
trial
The adjuvant potential of bpV(phen) was tested in vivo in the context of
Leishmania infection and resulted in complete protection against infectious
challenge.
In the past, use of total and soluble Leishmania antigen (SLA) for vaccination
has been reported and permitted some levels of protection against infectious
challenges (85). Using a murine model, we have tested whether bpV(phen)-
modulated cytokine and chemokine generation (as reported above) in
combination with SLA administration could lead to protection against
Leishmania infection. Mice have received PBS and bpV(phen) as controls,
and SLA (100 fig) with or without daily bpV(phen) injection (500 nM, 5 days).
All injections were done intra-peritoneally. Two weeks later, all groups were
inoculated similarly with their respective treatment. Finally, 4 weeks post-
vaccination, all animals were challenged with infectious Leishmania (5x106 L.
major intradermally injected in the right hind footpad) and progression of
infection followed over a period of 4 weeks post-infection (Fig. 10).
Protection
mediated by SLA+bpV(phen) has been successful since no significant

CA 02386759 2002-05-17
36
footpad inflammation and skin lesion development was observed in
comparison to all the other experimental groups.
E) PEROXOVANADIUM COMPOUNDS AND THE INHIBITION OF
TUMOR GROWTH
Methods
'- Cells
ZR-75: hormone-dependent cancer (ductal carcinoma) with oestrogen
receptors (ATCC, USA is depository)
PC-3: adenocarcinoma (grade IV) with bone metastasis (ATCC, USA is
depository).
A) ZR-75-1 human breast cancer: ZR-75-1 human breast cancer cells
obtained from the American Tissue Culture Collection (Rockville, MD) were
cultured in phenol red-free RPMI 1640. The cells were supplemented with
2mM L-glutamine, 1 mM Na-pyruvate, 100 IU penicillin/ml, 100~,g
streptomycin/ml, and 10% (v/v) fetal bovine serum and incubated under a
humidified atmosphere comprised of 95% air arid 5% COZ at 37°C.
Female homozygous HSD nu/nu athymic mice (50 days old) were obtained
from Harlan Sprague Dawley Inc. (Indianapolis, IN). Five mice were housed
per vinyl cage, which was equipped with air filter lids and kept in laminar
air
flow hoods under pathogen-limiting conditions. The photoperiod was
composed of a period of 14h of light and a period of 10h of darkness. Cages,
bedding, and food (Agway Pro-Lab R-M-H diet #4018) were autoclaved prior
to use. Water was acidified to pH of 2.8, autoclaved, and provided ad libitum.
Bilateral ovariectomy (OVX) was performed on all animals one week prior to
cell inoculation, under 2.5% isoflurane anesthesia mixed with oxygen.
Simultaneously, an oestrogen (E2) implant was inserted subcutaneously to

CA 02386759 2002-05-17
37
stimulate initital tumor growth and appearance. E2 implants were prepared in
1-cm long silastic tubing (inside diameter, 0.062 inch; outside diameter,
0.095
inch) containing 0.5 cm of estradiol/cholesterol diluted at a ratio of 1:10
(w:w).
One week after the ovariectomy, 2.0 x 106 ZR-75-1 cells, in their logarithmic
growth phase, were harvested with 0.083% pancreatin/0.3 mM EDTA and
inoculated s.c. in 0.1 ml of RPMI 1640 culture medium containing 30% of
Matrigel, from each flank of each animal through a 2.5-cm-long 20-gauge
needle. Four weeks after ZR-75-1 cell inoculation, the E2 implants were
replaced in all animals by estrone-containing implants (E~: cholesterol; 1:25
w:w) (104). Treatments consisting of increased doses of bpV(phen) versus
control were started 5 weeks after cell inoculation. Mice bearing tumors of an
average area of 15 mm2 were randomly assigned to 3 groups, each group
containing more than 15 mice. OVX animals first received the most potent
natural estrogen, to initiate cell proliferation and the development of
tumors.
Thereafter, the E2 implants were replaced by E~ implants as a model for post-
menopausal women in which E~ is the main circulating estrogen that is
converted into E2 in peripheral tissues.
On day 0 of the experiment (5 weeks after inoculation), the E~-releasing
implants were removed from the animals in Group 1 only. All mice received a
daily administration of bpV(phen) over a period of 42 days (i.p., 1001, in a 2
blind manner). Groups 1 and 2 received PBS, Group 3 received 2.5 mg/Kg
bpV(phen).
B) Human prostate adenocarcinoma (PC-3) in the athymic mice: Male
Balb/c nude (nu/nu) were purchased at 4-6 weeks of age from Charles Rivers
Inc. Mice were housed under pathogen free conditions and maintained on a
12-h light/12-h dark cycle with food and water supplied ad libitum. The
hormono-independent PC3 human tumor cells were from the American
Tissue Cuture Collection. Cells were grown in DMEM in the presence of 5
foetal bovine serum. Cells were collected at confluence, included in a matrix

CA 02386759 2002-05-17
38
(1.0 X 106 cells /ml; 30 % Matrigel). An equal volume of the tumor cell
suspension was injected s.c. in the right flank of each mouse. After 5 days, a
palpable tumor of approximately 5X5 mm was detected in the inoculated
animals. Mice with palpable tumors were divided into five groups (18
mice/group) for the treatment study. All mice in each treatment group had
tumor of similar size at the start of treatment. For administration to mice,
bpV(phen) was dissolved and diluted in phosphate buffered saline (PBS) at
pH 7.4. A Smg/Kg dose of bpV(phen) was administed daily by i.p. for 39
days. Taxol was used as a positive control at the dose of 20mg/Kg and
injected i.p. at every three days. A control group of 10 animals was injected
with PBS. The injection volume was kept constant at 100 pl/g body weight.
The mice were weighed three times during the experimental period to assess
toxicity of the treatment, and the tumors were measured twice weekly using
calipers. Tumor volume was calculated from the two-dimensional caliper
measurements using the following formula: tumor volume = length X (width)2
X 0.53.
The treatment period was completed after 39 days, when the PBS treated
group of mice had large tumors, requiring that the animals be sacrificed
according to the Animal Care Procedures. On the final day of the study, the
mice were sacrificed by carbon dioxyde inhalation. The s.c. tumors was
removed and weighed.
Statistical analysis: Tumor growth curves are presented in terms of
treatment group means and SEs. Statistical significance of treatment effect
was assessed by repeated measures ANOVA after applying a power
transformation to equalize residual variances and linearize the tumor growth
curves.

CA 02386759 2002-05-17
39
Results
1. Progression of tumor cells in vitro
The cells embedded in the collagen gel, grew as a "primary tumor". Some
cells migrated from the primary tumor towards the fibrin gel, forming front
edges. Small clumps of cells were observed in the fibrin gel; in this model
they represent "secondary tumors". Their extension and numbers are
representative of the invasive potential of the cancer cells.
PC-3: In control experiments, PC-3 cells migrated slightly from the primary
tumor and formed extensive secondary tumors in the fibrin gel. In the
presence of 2pM bpV(phen), a decrease in the size of the secondary tumor
was observed and the migration front from the primary tumor was similar to
1 S that seen in the control gels. In the presence of 5 and 10 uM bpV(phen),
there was no migration front and there were no secondary tumors in the fibrin
gel. In addition, the primary tumors appeared clearer than in the control
(Figure 11 ).
ZR-75-1: In control experiments ZR-75-1 cells migrated into the fibrin as
small spheroidal secondary tumors, with a limited and sparsely visible
migration front. The presence of 2pM bpV(phen) restricted the growth of the
primary tumor. At 5 and 10~M bpV(phen) there were no secondary tumors
and the primary tumor had a lower cell density (Figure 12).
2. Inhibition of tumor progression in vivo
A- ZR-75-1 human breast cancer:
The tumor size in the control group, having not received E~ replacement
therapy, did not increase. The tumor size in the animals in the other control
groups having received E~ replacement therapy was found to have increased

CA 02386759 2002-05-17
significantly (p<0.05) from 15 to 26 mm2 on day 42. The daily administration
of bpV(phen) do not resulted in increase of tumor size (p<0.05). The results
show that bpV(phen) has the capacity to inhibit the progression of tumors in
vivo (Figure 13A).
5
B- Human prostate adenocarcinoma (PC-3) in the athymic mice'
Daily administration of bpV(phen) caused a significant (p<0.001 ) 59
suppression of the final tumor compared with PBS-treated control animals
10 (Figure 13B ). No death were observed among the vehicle-treated controls or
bpV(phen), and, on average these mice gained 1.5 and 1.7 grams in body
weight respectively, relative to their weight at the initiation of the
treatment.
15 F) THE USE OF INHIBITORS OF PROTEIN TYROSINE PHOSPHATASES
(PTP) FOR ANTI-TUMOR IMMUNOTHERAPY
Lymphocytes with anti-tumor activity can be isolated from patients and grown
in vitro for use in cell-tranfer therapies (119). The incubation of immune
cells
20 with the PTP inhibitor bpV(phen) augments their activation state (120).
Therefore, the re-administration of bpV(phen)-activated immune cells to
cancer patients may enhance the immune response towards tumor cells. The
results described below demonstrate the efficacy of a method in which a
peroxometallic compound (bpV(phen) is used ex vivo on autologous immune
25 cells in order to stimulate the potency of these cells and once returned
into
blood circulation of cancer patients fight invasion malignant cells.
Method
30 An in vitro cancer invasion system that has been previously designed was
used. Briefly, prostate cancer cells (PC-3; American Type Culture Collection,
Rockville MD) were grown in DMEM medium with 5 % fetal bovine serum, 2
mM L-glutamine, and antibiotics. They were, incubated under a humidified
atmosphere of 95 % air/5% C02 at 37~C. Collagen gels containing the PC-3

CA 02386759 2002-05-17
41
cells were prepared according the method of Esdale and Bard (121 ). The
cell-embedded collagen gels were laid down onto a layer of fibrin gel, and
anchored by a second layer of fibrin gel. The top of the collagen gel was not
fully covered with fibrin gel in order to allow direct contacts between the
cancer cells in collagen and the splenocytes. The latter were directly seeded
onto the top layer of the collagen and fibrin gel. Prior to the molding of the
cancer invasion system, leucocytes were isolated from spleen of either
healthy mice or mice bearing PC-3 tumors. They were treated in vitro with
bpV(phen) (25 NM) for 24 hr. Thereafter, treated cells were washed, counted
and seeded (106 cells per gel) on the cancer cells-embedded gels. Untreated
leukocytes seeded on the top of the cancer invasion system (same
concentration) were used as a control experiment. Medium was renewed
periodically. During the whole experiment, most leucocytes remained on the
top of the gels, and have a normal morphology. Cell behavior was
periodically observed for 7 days of culture, then recorded (by photography).
Results
Clumps of PC3 cells progressively appeared in the fibrin gel representing the
invasive potential of the cancer cells. In the control 3D culture system, PC-3
cells migrated slightly from the primary site and formed extensive secondary
tumors in the fibrin gel as described in previous studies (122, 123). In
contrast to this, in the presence of bpV(phen)-treated leucocytes, neither
secondary tumors nor migration front was observed. In addition, the primary
tumors appeared less dense than in the control, indicating a smaller number
of growing cells (Figure 14).
Conclusion
The above describes a method consisting in the ex vivo autologous activation
by bpV(phen) of leucocytes and their potential to trigger a cellular immune
response against cancer cells. This may prove to be beneficial for several ex
vivo technologies. For example: alone or in combination with interleukins and

CA 02386759 2002-05-17
42
chemokines that trigger immune response to specific antigens or mutated cell
types (124).
Although the present invention has been described by way of preferred
embodiments thereof, these embodiments can be modified at will, within the
scope of the appended claims, without departing from the spirit and nature of
the subject invention.

CA 02386759 2002-05-17
43
LIST OF REFERENCES
1. Posner, B.I., Faure, R., Burgess, J.W., Bevan, A.P., Lachance, D.,
Zhan-Sun, G., Fantus, I.G., Ng, J.B., Hall, D.A., Lum, B.S. & Shaver,
A. (1994). Peroxovanadium compounds: a new class of potent
phosphotyrosine-phosphatase inhibitors which are insulin mimetics. J.
Biol. Chem. 269: 4596-4604.
2. Faure, R., Vincent, M., Dufour, M., Shaver, A. & Posner, B. (1995).
Arrest at the G2/M transition of the cell-cycle by protein-tyrosine
phosphatase inhibition: Studies on a neuronal and a glial cell line. J.
Cell. Biochem. 58: 389-401.
3. Tonks, N.K. and B.G. Heel Combinatorial control of the specificity of
PTPs. Current opinion in cell Biology 2001 13(2): 182-195.
4. Huyer et al. (1997)
5. Reydi, M. A. & Schwartz, S. M. (1981 ) Endothelial regeneration: Time
course of initial changes after small defined injury to rat aortic
endothelium. Lab. Invest. 44: 301-308.
6. Schwartz, S. M. & Liaw, L. (1993) Growth control and morphogenesis
in the development of pathology of arteries. J. Cardiovasc. Pharmacol.
21 (supply: S31-S49.
7. Auerback, R. (1994). Angiogenesis inhibition: a review. Pharmac.
Ther. 63: 265-311.
8. Folkman, J. (1995). Angiogenesis in cancer, vascular, rhumatoid and
other diseases. Nature Med. 1: 27-31.
9. Schreiber, A.B., Winkler, M.E. & Derynck, R. (1986). Transforming
growth factor-alpha: a more potent angiogenic factor than epidermal
growth factor. Science 232: 1250-1254.
10. Roberts, A.B., Sporn, M.B., Assoian, R.K., Smith, J.M., Roche, N.J. &
Wakefield, L.M. (1986). Transforming growth factor type beta: rapid
induction of fibrosis and angiogenesis in vivo and stimulation of
collagen formation in vitro. Proc. Natl. Acad. Sci. USA 84: 4167-4171.

CA 02386759 2002-05-17
44
11. Frater-Schroder, M., Risau, W., Hallmann, R., Gautschi, R. & Bohlen,
P. (1987). Tumor necrosis factor type-alpha, a potent inhibitor of
endothelial cell growth in vifro, is angiogenic in vivo. Proc. Natl. Acad.
Sci. USA 84: 5277-5282.
12. Fett, J.W., Strydom, D.J., Lobb, R.R., Alderman, R.M., Riordn, J.F. &
Vallee, B.L. (1985). Isolation and characterization of angiogenin, an
angiogenic protein from human carcinoma cells. Biochemistry 24:
5480-5488.
13. Ziche, M., Jones, J. & Gullino, P. (1982). Role of prostaglandin E1 and
copper in angiogenesis. J. Natl. Cancer Inst. 69: 475-482.
14. Dobson, D.E., Kambe, A., Block, E., Dion, T., Lu, H. 8~ Castellot, Jr.
J.J., Speigelman, B.M. (1990). 1-Butyrylglycerol: a novel angiogenesis
factor secreted by differentiating adipocytes. Cell 61: 223-230.
15. Baird, A. & Durkin, T. (1986). Inhibition of endothelial cell
proliferation
by type-beta transforming growth factor: interaction with acidic and
basic fibroblast growth factors. Biochem. Biophys. Res. Commun. 138:
476-482.
16. Frater-Schroder, M., Muller, G., Birchmeier, W. & Bohlen, .P. (1986).
Transforming growth factor-beta inhibits endothelial cell proliferation.
Biochem. Biophys. Res. Commun. 137: 295-302.
17. Folkman, J., & Klagsbrun, M. (1987). Angiogenic factors. Science 235:
442-447.
18. Klagsbrun, M. & D'Amore, P.A. (1991 ). Regulators of angiogenesis.
Annu. Rev. Physiol. 53: 217-232.
19. Gospodarowicz, D., Ferrara, N., Schweigerer, L. & Neufeld, G. (1987).
Structural characterization and biological functions of fibroblast growth
factor. Endocr. Rev. 8: 95-104.
20. Thomas, K., Rios Candelore, M., Gimenez-Gallego, G., Di Salvo, J.,
Bennet, C., Rodkey, J. & Fizpatrick, S. (1985). Pure brain-derived
acidic fibroblast growth factor is a potent vascular endothelial cell
mitogen with sequence homology to interleukin-1. Proc. Nat!. Acad.
Sci. USA 82: 6409-6416.

CA 02386759 2002-05-17
21. Miyazono, K., Okabe, T., Urabe, A., Takata, F. & Heldin, C.-H. (1987).
Purification and properties of an endothelial cell mitogen from human
platelets. J. Biol. Chem. 262: 4098-4104.
22. Ishikawa, F., Miyazono, K., Hellman, U., Drexler, H., Westerndt, C.,
5 Hagiwara, K., Usuki, K., Takaku, F., Risau, W. & Heldin, C.-H. (1989).
Identification of angiogenic activity and the cloning and expression of
platelet-derived endothelial cell growth factor. Nature 338: 557-561.
23. Leung, D.W., Cachianes, G., Kuang, W.-J., Goeddel, D.V. & Ferrara,
N. (1989). Vascular endothelial growth factor is a secreted angiogenic
10 mitogen. Science 246: 1306-1312.
24. Houck, K.A., Ferrara, N., Winer, J., Cachianes, G., Li, B. & Leung,
D.W. (1991 ). The vascular endothelial growth factor family:
Identification of a fourth molecular species and characterization of
alternative splicing of RNA. Mol. Endocrinol. 5: 1806-1814.
15 25. Breier, G., Albrecht, U., Sterrer, S. & Risau, W. (1992). Expression of
vascular endothelial growth factor during embryonic angiogenesis and
endothelial cell differentiation. Development 114: 521-532.
26. Bussolino, F., Albini, A., Garnussi, G., Presta, M., Viglietto, G., Ziche,
M. & Persico, G. (1996). Role of soluble mediators in angiogenesis.
20 Eur. J. Cancer 32A: 2401-2412.
27. Colville-Nash, P.R. & Willoughby, D.A. (1997). Growth factors in
angiogenesis: current interest and therapeutic potential. Mol. Med.
Today. 3: 14-23.
28. Knighton, D.R., Hunt, T.K., Scheuenstuhl, H., Halliday, B.J., Werb, Z.
25 & Banda, M.J. (1983): Oxygen tension regulates the expression of
angiogenesis factor by macrophages. Science 221: 1283-1285.
29. Ladoux, A. & Frelin, C. (1993). Hypoxia is a strong inducer of vascular
endothelial growth factor mRNA expression in the heart. Biochem.
Biophys. Res. Commun. 195: 1005-1010.
30 30. Nomura, M., Yamagishi, S.I., Harada, S.I., Hayashi Y., Yarnashima,
T., Yamashita, J. & Yamamoto H. (1995). Possible participation of
autocrine and paracrine vascular endothelial growth factors in

CA 02386759 2002-05-17
46
hypoxia-induced proliferation of endothelial cells and pericytes. J. Biol.
Chem. 270: 28316-28324.
31. Walgenbach, K.J., Gratas, C., Shestak, K.C. & Becker, D. (1995).
Ischaemia-induced expression of bFGF in normal skeletal muscle: a
potential paracrine mechanism for mediating angiogenesis in
ischaemic skeletal muscle. Nature Med. 1: 453-459.
32. Lew, A.P., Lew, N.S., Iliopoulos, O., Jiang, C., Kaeling Jr., W.G. &
Goldberg, M.A. (1997). Regulation of vascular endothelial growth
factor by hypoxia and its modulation by the von Hippel-Lindau tumor
suppressor gene. Kidney Int. 51: 575-578.
33. Sandner, P., Wolf, K., Bergmaier, U., Gess, B. & Kurtz, A. (1997).
Induction of VEGF and VEGF receptor gene expression by hypoxia:
Divergent regulation in vivo and in vitro. Kidney Int. 51: 448-453.
34. Ferrara, N. (1995). Missing link in angiogenesis. Nature 376: 467.
35. Koch, A.E., Halloran, M.M., Haskell, C.J., Shaw, M.R. & Polverini, P.J.
(1995). Angiogenesis mediated by soluble forms of E-selectin and
vascular cell adhesion molecule-1. Nature 376: 517-519.
36. Polverini, J.P., Cotran, R.S., Gimbrone Jr, M.A. & Unanue, E.R.
(1977). Activated macrophages induce vascular proliferation. Nature
269:804-806.
37. Sunderkotter, C., Steinbrink, K., Goebeler, M., Bhardwaj, R. & Sorg,
C. (1994). Macrophages and angiogenesis. J. Leukocyte Biol. 55: 410-
422.
38. Polverini, P.J. (1996). How the extracellular matrix & macrophages
contribute to angiogenesis-dependant diseases. Eur. J. Cancer 14:
2430-2437.
39. Gross, J.L., Moscatelli, D., Jaffe, E.A. & Rifkin, D.B. (1982).
Plasminogen activator and collagenase production by cultured
capillary endothelial cells. J. Cell Biol. 95: 974-981.
40. Moscatelli, D.A., Rifkin; D.B. & Jaffe, E.A. (1985). Production of latent
collagenase by human umbilical vein endothelial cells in response to
angiogenic preparations. Exp. Cell Res. 156: 379-390.

CA 02386759 2002-05-17
47
41. Ingber, D.E. (1991 ). Extracellular matrix and cell shape: potential
control points for inhibition of angiogenesis. J. Cell. Biochem. 47: 236-
241.
42. Ingber, D.E. (1992). Extracellular matrix as a solid-state regulator in
angiogenesis: identification of new targets for anti-cancer therapy.
Sem. Cancer Biol. 3: 57-63.
43. Bischoff, J. (1995). Approaches to studying cell adhesion molecules in
angiogenesis. Trends Cell Biol. 5: 69-74.
44. Eisenstein, R. (1991 ). Angiogenesis in arteries: review. Pharmac.
Ther.49:1-19.
45. Folkman, J. & Ingber, D. (1992). Inhibition of angiogenesis. Cancer
Biol. 3: 89-96.
46. Bicknell, R. (1994). Vascular targeting and the inhibition of
angiogenesis. Ann. Oncol. 5: S45-S50.
47. Montesano, R., Pepper, M.S., Belin, D., Vassalli, J.-D. & Orci, L.
(1988). Induction of angiogenesis in vitro by vanadate, an inhibitor of
phosphotyrosine phosphatases. J. Cell. Physiol. 134: 460-466.
48. Montesano, R. & Orci, L. (1985). Tumor-promoting phorbol esters
induce angiogenesis in vitro. Cell 42: 469-477.
49. Montesano, R., Vassalli, J.D., Baird, A., Guillemin, R. & Orci, L.
(1986). Basic fibroblast growth factor induces angiogenesis in vitro.
Proc. Natl. Acad. Sci. USA. 83: 7297-7301.
50. Sefton, B.M. & Hunter, T. (1984). Tyrosine protein kinases. Adv. Cyclic
Nucleotide Protein Phosphorylation Res. 18: 195-203.
51. Hunter, T. & Cooper, J.A. (1985). Protein-tyrosine kinases. Annu. Rev.
Biochem. 54: 897-930.
52. Hunter, T. & Cooper, J.A. (1986). Viral oncogenes and tyrosine
phosphorylation. In: The Enzymes, Vol. 17. P.D. Boyer and E.G.
Krebs, eds. Academic Press, Orlando, Florida, pp. 191 -246.
53. Ramachandran, J. & Ullrich, A. (1987). Hormonal regulation of protein
tyrosine kinase activity. Trends Pharmacol. Sci. 8: 28-31.

CA 02386759 2002-05-17
48
54. Battistini, B., Chailler, P., D'Orleans, J., Briere, N. & Sirois, P.
(1993).
Growth regulatory properties of endothelins. Peptides 14: 385-399.
55. Pedram, A., Razandi, M., Hu R-M & Levin, E.R. (1997). Vasoactive
peptides modulate vascular endothelial cell growth factor production and
endothelial cell proliferation and invasion. J. Biol. Chem. 272: 17097-
17013.
56. Asham, E.H., Loizidou, M. & Taylor, I. (1998). Endothelia-1 and tumor
development. Eur. J. Surg. Oncol. 24: 57-59.
57. Kusuhara, M., Yamaguchi, K., Nagasaki, K., Hayashi, C., Suzaki, A.,
Nori, S., Hanada, S., Nakamura, Y. and Abe, K. (1990). Production of
endothelia in human cancer cell lines. Cancer Res. 50: 3257-3261.
58. Schichiri, M., Hirata, Y., Nakajiroa, T., & o, K., Imal, T., Yanagisawa,
M., Mazaki, T. and Marumo, F. (1991 ). Endothelia-I is an
autocrine/paracrine growth factor for human cancer cell lines. J. Clin.
Invest. 87: 1867- 1871.
59. Pagotto, U., Arzberger, T., Hopfer, U., Weidnl, A. & Stella, G. (1995).
Cellular localization of endothelia receptor mRNA ETA and ETg in the
brain tumor and normal human brain. J. Cardiovasc. Pharmacol. 26
(Suppl. 3): S104-106.
60. Stiles, J.D., Ostrow, P.T., Balos, L.L., Greenberg, S.J., Plunkett, R.,
Grand, W. & Heffner Jr., R.R. (1997). Correlation of endothelia-I and
transforming growth factor X31 with malignancy and vascularity in
human gliomas. J. Neurobiol. 56: 435-439.
61. Ishibashi, M., Fujita, M., Nagai, K., Koko, M., Furue, H., Haku, E.,
Osamura, Y. & Yamaji, T. (1993). Production and secretion of
endothelia by hepatocellular carcinoma. J. Clin. Endocrinol. Metab. 76:
378-383.
62. Giaid, A., Hamid, Q.A., Springall, D.R., Yanagisawa, M., Shinmi, O.,
Sawamura, T., Masaki, T., Kimura, S., Corrin, B. and Polak, J.M.
(1990). Detection of endothelia immunoreactivity and mRNA in
pulmonary tumors. J. Pathol. 162: 15-22.

CA 02386759 2002-05-17
49
63. Nelson, J., Chan, K., Hedican, S., Magnuson, S., Opgenorth, T., Bova,
G. & Simons, J. (1996). Endothelin- 1 production decreased B
receptor expression in advanced prostate cancer. Cancer Res. 56:
663-668.
64. Kikuchik, K., Nakagawa, H., Kadono, T., Etoh, T., Byers, H., Mihm, M.
& Tamaki, K. (1996). Decreased ETB receptor expression in human
metastatic melanoma cells. Biochem. Biophys. Res. Commun. 219:
734-739.
65. Ziche, M., Morbidelli, L., Donnini, S. & Ledda, F. (1995). ETB receptor
promote proliferation and migration of endothelial cells. J. Cardiovasc.
Pharmacol. 26 (Suppl. 3): S284-S286.
66. Inagaki, H., Bishop, A., Escrig, C., Wharton, J., Allen-Mersh, T. &
Polak, J. (1991 ). Localization of endothelin like immunoreactivity and
endothelin binding sites in human colon. Gastroenterology 101: 47-54.
67. Inagaki, H., Bishop, A., Eimoto, T. & Polak, J. (1992).
Autoradiographic localization of endothelin-1 binding sites in human
colonic cancer tissue. J. Pathol. 168: 263-267.
68. Eguchi, S., Hirata, Y., Imai, T. & Marumo, P. (1995). ET-1 as an
autocrine growth factor for endothelial cells. J. Cardiovasc. Pharmacol.
26 (Suppl. 3): S279-S283.
69. Leveson, S., Wiggins, P., Giles, G., Parkin, A. & Robinson, P. (1985).
Deranged blood flow patterns in the detection of liver metastases. Br.
J. Surg. 72: 128-130.
70. Carter, R., Anderson, J., Cooke, T., Baxter, J. & Angerson, W. (1994).
Splanchnic blood flow changes in the presence of hepatic tumors,
evidence of humoral mediator. Br. J. Cancer 69: 1025-1026.
71. Nakamura, T., Ebihara, I., Fukui, M., Tomino, Y. & Koide, H. (1995).
Effect of a specific endothelin receptor A antagonist on mRNA levels
for extracellular matrix components and growth factors in diabetic
glomeruli. Diabetes 44: 895-899.

CA 02386759 2002-05-17
72. Lee, K.M., Tsai, K.Y., Wang, N. & Ingber, D.E. (1998). Extracellular
matrix and pulmonary hypertension: control of vascular smooth
muscle cell contractility. Am. J. Physiol. 274: H76-82.
73. Hirshfeld, J.W. Jr, Schwartz, J.S., Jugo, R., MacDonald, R.G.,
5 Goldberg, S., Savage, M.P., Bass, T.A., Vetrovec, G., Cowley, M.,
Taussig, A.S., et al. (1991) Restenosis after coronary angioplasty: a
multivariate statistical model to relate lesion and procedure variables
to restenosis. The M-HEART Investigators. J. Am. Coll. Cardiol. 18:
647-656.
10 74. Maseri, A. (1996). An unresolved, persisting challenge for
percutaneous transluminal coronary angioplasty: how to identify the
lesions that will not restenose. Eur. Heart J. 17: 814-815.
75. Kirchengast, M. & Munter, K. (1998). Endothelia and restenosis.
Cardiovasc. Res. 39: 550-555.
15 76. Kramer, B.K., Ackermann, M., Kohler, S.M. & Riegger, G.A. (1994).
Role of endothelia in hypertension. Clin. Investig. 72: 88-93.
77. Levin, E. R. (1995). Endothelins: mechanisms of disease. New Eng. J.
Med. 333: 356-363.
78. Rohmeiss, P., Birck, R., Braun, C., Kirchengast, M. & van der Woude,
20 F.J. (1999). Targets for endothelia in the diseased kidney: clues for
therapeutic intervention. Exp. Nephrol. 7: 1-10.
79. Cardell, L.O., Uddman, R. & Edvinsson, L. (1994). Endothelins: a role
in cerebrovascular disease? Cephalalgia 14: 259-65.
80. Cosentino, F. & Katusic, Z. S. (1994). Does endothelia-1 play a role in
25 the pathogenesis of cerebral vasospasm? Stroke 25: 904-908.
81. Battistini, B. & Dussault, P. (1998). The many aspects of endothelins
in ischemia-reperfusion injury: emergence of a key mediator. J. Invest.
Surg. 11: 297-313.
82. Mauel, J. (1990). Macrophage-parasite interactions in Leishmania
30 infections. J. Leukoc. Bioi. 47: 187-193.

CA 02386759 2002-05-17
51
83. Belosevic, M., Findbfoom, D.S., Meltzer, M.S. & Nacy, C.A. (1990). IL-
2. A cofactor for induction of activated macrophage resistance to
infection. J. Immunol. 145: 831-839.
84. Olivier, M., Bertrand, S. & Tanner, C. E. (1989). Killing of Leishmania
donovani by activated liver macrophages from resistant and
susceptible strains mice. lnt. J. Parasitol. 19: 377-383 .
85. Skov, C.B. & Twohy, W. (1974). Cellular immunity to Leishmania
donovani. I. The effect of T cell depletion on resistance to L. donovani
in mice. J. Immunol. 113: 2004-2011.
86. Murray, H.W., Masur, H. 8~ Keithly, J.S. (1982). Cell-mediated
immune response in experimental visceral leishmaniasis. I. Correlation
between resistance to Leishmania donovani and lymphokine-
generating capacity. J. Immunol. 129: 344-350.
87. Olivier, M. & Tanner, C. (1989). The effect of cyclosporin A in murine
visceral leishmaniasis. Trop. Med. Parasitol. 40: 32-38.
88. Refiner, N.E. & Finke, J.H. (1983). Interleukin 2 deficiency in murine
leishmanfiasis donovani and its relationship to depressed spleen cell
responsees to phytohemagglutinin. J. Immunol. 131: 1487-1491.
89. Murray, H.W., Stern, J.J., Welte, K., Rubin, B.Y., Carriero, S.M. &
Nathan, C.F. (1987). Experimental visceral leishmaniasis: production
of interleukin 2 and interferon-gamma, tissue immune reaction, and
response to treatment with interleukin 2 and interferon-gamma. J.
Immunol. 138: 2290-2297.
90. Olivier, M., Proulx, C. & Tanner, C.E. (1989). Importance of
lymphokines in the control of the multiplication and dispersion of
Leishmania donovani within liver macrophages of resistant and
susceptible mice. J. Parasitol. 75: 720-727.
91. Green, S.P., Hamilton, J.A. & Phillips, W.A. (1992). Zymosan-triggered
tyrosine phosphorylation in mouse bone-marrow-derived
macrophages is enhanced by respiratory-burst priming agents.
Biochem. J. 288: 427-432.

CA 02386759 2002-05-17
52
92. Greenberg, S., Chang. P. & Silverstein, S.C. (1993). Tyrosine
phosphorylation of the gamma subunit of Fc gamma receptors,
p72syk, and paxillin during Fc receptor-mediated phagocytosis in
macrophages. J. Biol. Chem. 269: 3897-3902.
93. Dong, Z., Qi, X., Xie, K. & Fidler, I.J. (1993). Protein tyrosine kinase
inhibitors decrease induction of nitric oxide synthase activity in
lipopolysaccharide-responsive and lipopolysaccharide-nonresponsive
murine macrohages. J. Immunol. 151: 2717-2724.
94. Dong, Z., O'Brian, C.A. & Fidler (1993). 1.J., Activation of tumoricidai
properties in macrophages by lipopolysaccharide requires protein-
tyrosine kinase activity. J. Leukoc. Biol. 53: 53-60.
95. Glaser, K.B., Sung, A., Bauer, J. & Weichman, B.M. (1993).
Regulation of eicosanoid biosynthesis in the macrophage. Involvement
of protein tyrosine phosphorylation and modulation by selective protein
tyrosine kinase inhibitors. Biochem. Pharmacol. 45: 711-721.
96. Golden, A., Nemeth, S.P. & Brugge, J.S. (1986). Blood platelets
express high levels of the pp60c-src-specific tyrosine kinase activity.
Proc. Natl. Acad. Sci. USA 83: 852-856.
97. Golden, A. & Brugge, J.S. (1989). Thrombin treatment induces rapid
changes in tyrosine phosphorylation in platelets. Proc. Natl. Acad. Sci.
USA 86: 901-905.
98. Olivier, M, Romero-Gallo B. J., St-Laurent, R., Racette, N., Stevenson,
M., Jacobs, P., Posner, B.L, Tremblay, M., & Faure, R. (1998).
Modulation of INFg induced macrophage activation by
phosphotyrosine phosphatase inhibition. J. Biol. Chem. 273: 13944-
13949.
99. Chao, W., Liu, H., Hanahan, D.J. 8~ Olson, M.S. (1992). Protein
tyrosine phosphorylation and regulation of the receptor for platelet-
activating factor in rat Kupffer cells. Effect of sodium vanadate.
Biochem. J. 288: 777-784.

CA 02386759 2002-05-17
53
100. Secrist, J.P., Burns, L.A., Karnitz, L., Koretsky, G.A. & Abraham, R.T.
(1993). Stimulatory effects of the protein tyrosine phosphatase
inhibitor, pervanadate, on T-cell activation events. J. Biol. Chem. 268:
5886-5893.
101. Imbert, V., Peyron, J.F., Farahi Far, D., Mari, B., Auberger, P. & Rossi,
B. (1994). Induction of tyrosine phosphorylation and T-cell activation
by vanadate peroxide, an inhibitor of protein tyrosine phosphatase.
Biochem. J. 297: 163-173.
102. Barbeau, B., Bernier, R., Dumais, N., Briand, G., Olivier, M., Faure,
14 R.,. Posner, B.I. & Tremblay, M. (1997). Activation of HIV-1 LTR
transcription and virus replication via NF-OB-dependent and -
independent pathways by potent phosphotyrosine phosphatase
inhibitors, the peroxovanadium compounds. J. Biol. Chem. 272:
12968-12977.
103. Janvier, R., Sourla, A., Koutsilieris, M. & Doillon, C. (1997). Stromal
Fibroblasts are required for PC-3 human prostate cancer cells to
produce capillary-like formation of endothelial cells in a three-
dimensional co-culture system. Anticancer res. 17: 1551-1558.
104. Couillard S, Gutman M, Labrie F, Candos B, Labrie C. (1999). Effect
of combined treatment with EM-800 and radiotherapy on the growth of
human 212-75-1 breast cancer xenografts in nude mice. Cancer
Res.59: 4857-4863.
105. Sirois, E., Cots, M.F. & Doillon, C.J. (1993). Growth factors and
biological supports for endothelial cell lining: in vitro study. Int. J.
Artificial Organs 16: 609-619.
106. Nicosia, R.F. & Ottineti, A. (1990) Growth of microvessels in serum-
free-matrix culture of aorta. A quantitative assay of angiogenesis in
vitro. Lab. Invest., 63:115-122.
107. Passaniti, A., Taylor, R.M., Pili, R., Guo, Y., Long, P.V., Haney, J.A.,
Pauly, R.R., Grant, D.S. & Martin, G.R. (1992). A simple quantitative
method for assessing angiogenesis and antiangiogenic agents using
reconstituted basement membrane, heparin, and fibroblast growth

CA 02386759 2002-05-17
54
factor. Lab. Invest. 67 : 519-528.
108. Laporte, S., & Escher, E. (1992). Neointirna formation after vascular
injury is angiotensin II mediated. Biochem. Biophys. Res. Comm.
181:1510-1516.
109. Lima, G.M., Vallochi, A.L., Sllva, U.R., Bevilacqua, E.M., Kiffer, M.M. &
Abrahamsohn, I.A. (1998). The role of polymorphonuclear leukocytes
in the resistance to cutaneous leishmaniasis. Immunol. Lett. 64: 145-
151.
110. Hobbs, A.J., Higgs, A. & Moncada, S. (1999). Inhibition of nitric oxide
synthase as a potential therapeutic target. Annu. Rev. Pharmacol.
Toxicol. 39: 191-220.
111. Ajuevor, M.N., Virag, L., Flower, R.J., Perretti, M. & Szabo, C. (1998).
Role of inducible nitric oxide synthase in the regulation of neutrophil
migration in zymosan-induced inflammation. Immunology. 95: 625
630.
112. Yan, X.T., Tumpey, T.M., Kunkel, S.L., Oakes, J.E. & Lausch, R.N.
(1998). Role of MIP-2 in neutrophil migration and tissue injury in the
herpes simplex virus-1-infected cornea. Invest. Ophthalmol. Vis. Sci.
39: 1854-1862.
113. Duong, M., Ouellet, N., Simard, M., Bergeron, Y., Olivier, M. &
Bergeron M.G.B. (1998). Kinetic study of host defense and
inflammatory response to Aspergillus fumigatus in steroid-induced
immunosuppressed mice. J. Infect. Dis. 178: 1472-1482.
114. Tam, F.W., Karkar, A.M., Smith, J., Yoshimura, T., Steinkasserer,
~Kurrle, R., Langner, K. & Rees, A.J. (1996) Differential expression of
macrophage inflammatory protein-2 and monocyte chemoattractant
protein-1 in experimental glomerulonephritis. Kidney Int. 49: 715-721.
115. Stenger, S., Thiiring, H., Rollinghoff, M. & Bogdan, C. (1994). Tissue
expression of inducible nitric oxide synthase is closely associated with
resistance to Leishmania major. J. Exp. Med. 180: 783-793.

CA 02386759 2002-05-17
116. Stenger, S., Donhauser, N., Thuring, H., Rollinghoff, M. & Bogdan, C.
(1996). Reactivation of latent leishmaniasis by inhibition of inducible
nitric oxide synthase. J. Exp. Med. 183: 1501-1514.
117. Pani, G., Kozlowski, M., Cambier, J., Mills, G.B. & Siminovitch, K.A.
5 (1995) Identification of the tyrosine phosphatase PTP1C as a B cell
antigen receptor-associated protein involved in the regulation of B cell
signaling. J. Exp. Med.. 181: 2077-2084.
118. Klingmiiller, U., Lorenz, U., Cantley, L.C., Neel, B.G. & Lodish, H.F.
(1995). Specific recruitment of SH-PTP1 to the erythropoietin receptor
10 causes inactivation of JAK2 and termination of proliferative signals.
Cell 80: 729-738.
119. Rosenberg, S.A. Progress in human tumor immunology and
immunotherapy. Nature 411: 380-384. 2001.
120. Olivier, M. Romero-Gallo B. J., St Laurent, R., Racette, N., Stevenson,
15 M., Jacobs, P., Posner, B., Tremblay, M., Faure, R. Modulation of
INFg-induced macrophage activation by phosphotyrosine
phosphatase inhibition. J. Biol. Chem. 273, 13944-13949 (1998).
121. Esdale, T., Bard, J. Collagen substrata for studies on cell behavior. J.
Cell. Biol. 54: 626-637 (1972).
20 122. Janvier, R., Sourla, A., Koutsilieris, M., Doillon, C. Stromal
fibroblasts
are required for PC-3 human prostate cancer cells to produce capillary
-like formation of endothelial cells in a three-dimensional co-culture
system. Anticancer Res. 17: 1551-1557 (1997).
123. Dvorak, H.F., Harvey, V.S., Estrella, P., Brown, L.F., Mc Donagh, J.,
25 Dvorak A.M. Fibrin containing gels induce angiogenesis. Implications
for stroma generartion and wound healing. Lab Invest. 57: 673-686
(1987).
124. Eberlein, T.J., Rosenstein, M. & Rosenberg S.A. Regression of a
disseminated syngeneic solid tumor by systemic transfer of lymphoid
30 cells expanded in interleukin 2. J. Exp. Med. 56: 385-397 (1982).

Representative Drawing

Sorry, the representative drawing for patent document number 2386759 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
(22) Filed 2002-05-17
(41) Open to Public Inspection 2003-11-17
Dead Application 2007-05-17

Abandonment History

Abandonment Date Reason Reinstatement Date
2006-05-17 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $150.00 2002-05-17
Registration of a document - section 124 $100.00 2002-12-12
Registration of a document - section 124 $100.00 2002-12-12
Maintenance Fee - Application - New Act 2 2004-05-17 $50.00 2004-05-17
Maintenance Fee - Application - New Act 3 2005-05-17 $50.00 2005-05-02
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
UNIVERSITE LAVAL
MCGILL UNIVERSITY
Past Owners on Record
BATTISTINI, BRUNO JOSEPH
DOILLON, CHARLES
FAURE, ROBERT
OLIVIER, MARTIN
POSNER, BARRY
SAVARD, PIERRE
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) 
Description 2002-05-17 55 2,371
Abstract 2002-05-17 1 35
Cover Page 2003-10-21 1 41
Claims 2002-05-17 16 457
Correspondence 2002-06-27 1 24
Assignment 2002-05-17 3 101
Assignment 2002-12-12 6 232
Correspondence 2002-12-12 3 98
Assignment 2002-05-17 5 165
Fees 2004-05-17 1 36
Fees 2005-05-02 1 33
Drawings 2002-05-17 15 1,017