CA2297900A1 - Phosphonic acid naphthylureas and their antiantiogenic and antitumorigenic use - Google Patents

Phosphonic acid naphthylureas and their antiantiogenic and antitumorigenic use Download PDF

Info

Publication number
CA2297900A1
CA2297900A1 CA002297900A CA2297900A CA2297900A1 CA 2297900 A1 CA2297900 A1 CA 2297900A1 CA 002297900 A CA002297900 A CA 002297900A CA 2297900 A CA2297900 A CA 2297900A CA 2297900 A1 CA2297900 A1 CA 2297900A1
Authority
CA
Canada
Prior art keywords
suramin
substituted
phosphoric acid
acid
phosphonic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002297900A
Other languages
French (fr)
Inventor
Delwood C. Collins
Antonio R. Gagliardi
Peter Nickel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Kentucky Research Foundation
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/899,966 external-priority patent/US5831347A/en
Priority claimed from US09/121,124 external-priority patent/US6096730A/en
Application filed by Individual filed Critical Individual
Priority claimed from PCT/US1998/015470 external-priority patent/WO1999005148A1/en
Publication of CA2297900A1 publication Critical patent/CA2297900A1/en
Abandoned legal-status Critical Current

Links

Abstract

Novel phosphonic acid agents are synthesized and characterized and their pharmaceutical compositions provided. Their method of use for the inhibition of angiogenesis and metalloproteinase and the treatment of tumors is also shown. Embodiments of the phosphonic acid agents include: (P-Yn1)m1-Q1-K-(Q2- (Yn2-P)m2)j wherein P is a phosphonic group or a phosphonic salt, Y is -OCO-, NR1CO- or CON(R1)R2-; R1 is H, CH2CO2H, or substituted or unsubstituted alkyl; R2 is substituted alkyl, aryl or arylalkyl; Q1 and Q2 are substituted or unsubstituted aryl groups; K is H, -NH-CO-NH-, -NH-CS-NH-, -NHCO-R3-CONH- or NHCS-R3-CSNH-; provided when K is H, R3 is a substituted or unsubstituted aryl group; j is 0; j is 0,1,2; n1 and n2 are independently 0,1 or 2; and m1 and m2 are independently an integer from 1-4.

Description

1 _ The present invention relates to phosphoric acid agents that are potent inhibitors of angiogenesis and tumorigenesis.
This application claims priority under 35 U.S.C. ~ 120 from Serial No.
08/899,996, filed July 24, 1997.
Angiogenesis is an essential component of tumor growth and metastasis. As reviewed by Follcman (1985), the growth of solid tumors is dependent on angiogenesis.
Typically tumors do not grow beyond a size of 2-3 mm unless they are able to stimulate the growth of new capillaries from the existing vascular network. Additionally, the new blood vessels provide an essential entry route to the vasculature for metastasis of tumor cells. Cell division in endothelial cells is slow, with a turnover time of years rather than days or hours (Denekamp, 1984).
However, vascular endothelial cells undergo rapid proliferation with turnover times of a few days during the growth of new capillaries. Angiogenesis-dependent diseases such as diabetic retinopathy, psoriasis, arthritis, hemangiomas and tumor growth and metastasis are characterized by uncontrolled growth of capillary blood vessels. The most striking example of uncontrolled angiogenesis is associated with tumor growth (Folkman, 1985).
Accordingly, the search for angiogenesis inhibitors was stimulated by the concept of "antiangiogenic therapy". In its simplest terms, antiangiogenic therapy sought a putative inhibitor of blood vessel growth in the believe that such an inhibitor might be therapeutic by limiting tumor growth and further such an inhibitor would be non-toxic because angiogenesis is normally infrequent (Folkman, 1992). A number of different factors can stimulate angiogenesis in vivo. These angiogenic factors, such as basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and SUBSTtME SHEET (RULE 26) transforming growth factor a and Vii, can be released from the tumor cells themselves and by other cells such as macrophages and endothelial cells (Folkman, 1992).
The mechanisms by which tumors induce angiogenesis are very complex and involve many pathways, an angiogenesis inhibitor could be directed against any of the components of the angiogenic cascade. The identification of compounds that block neovascularization has a long standing interest. A number of inhibitory extracts have been prepared from avascular tissues, such as cartilage (Braunhut et al., 1989). One such method of treating tumors has been by the administration of suramin. However, it is believed that suramin may have adverse effects in large dosages.
Accordingly, a continuing need exists for agents that overcome the deficiencies of prior antiangiogenic compounds, including suramin. There is also a need for antiangiogenic agents that have a reduced toxicity to a recipient and increase inhibition of angiogenesis and tumorlgenesls.
It is an object of the present invention to provide novel phosphonic acid substituted agents and pharmaceutical compositions containing said agents.
In an embodiment, the invention provides methods of treating tumors comprising the steps of administering an effective amount of a phosphonic acid substituted agent to a patient in need of said treatment.
In another embodiment the invention provides a method of inhibiting angiogenesis comprising the steps of administering an effective amount of a phosphonic acid substituted agent to a patient in need of said treatment.
In satisfaction of the foregoing objects and advantages, the present invention provides phosphonic acid derivatives of agents and methods for their preparation.
The invention also provides pharmaceutical compositions and methods for use of the compositions as potent inhibitors ef angiogenesis and tumorigenesis.
In parent application Serial No. 08/889,996, the compounds of the invention were referred to as naphthylureas. However, the more appropriate class of the agents are as described herein.
The above and other objects of the invention will become readily apparent to those of skill in the relevant art from the following detailed description, tables and formulas, wherein SUBSTITUTE SHEET (RULE 26) only the preferred embodiments of the invention are shown and described, simply by way of illustration of the best mode of carrying out the invention.
The present invention provides a novel group of phosphoric acid agents which were synthesized and characterized. This unique group of compounds are potent inhibitors of angiogenesis, equipotent to 40 times greater than suramin. In addition, results show that phosphoric acid agents demonstrate lower toxicity and exert their antiangiogenic effect via a different mechanism than suramin.
Without subscribing to any particular theory, it is believed that phosphoric acid agents are potent inhibitors of angiogenesis and that the antiangiogenic effect is mediated through a specif c effect of these compounds upon proliferating endothelial cells. The mechanism for inhibition of angiogenesis by the phosphoric acid agents may involve inhibition of DNA
replication, cell signaling and/or energy production.
Furthermore, the antiangiogenic and endothelial cell growth inhibiting activity of the phosphoric acid agents is not primarily related to the inhibition of binding of the angiogenic growth factors to their receptors on the endothelial cell surface as has been demonstrated for suramm.
The findings that this unique group of phosphoric acid agents have more potent antiangiogenic activity, are less toxic than suramin, and are metabolized and cleared in hours rather than weeks suggest that these agents are potent therapeutic agents for "angiogenesis-dependent" diseases. These diseases include diabetic retinopathy, arthritis, psoriasis, tumor growth and metastasis.
The commercial advantage of the phosphoric acid agents resides is their lower toxicity and enhanced antiangiogenic activity. The experiments presented illustrate the potent inhibition of angiogenesis by the use of the phosphoric acid agents to inhibit the growth of various immortalized human cancer cell lines. Further experiments provide in vivo mouse toxicity studies and neurotoxicity in vitro assays. Additionally, animal studies are presented in which SCID or nude mice are implanted with human tumors and then treated with the selected phosphoric acid agents.
According to the present invention, phosphoric acid agents, described by structural formula and chemical name, are potent inhibitors of angiogenesis and/or tumorigenesis while exhibiting low toxicity.
SU6STlTUTE SHEET (RULE 26) The present invention provides a preferred novel class of phosphonic acid group substituted agents which are defined by the following formulae:
(P-Ym )ml- Ql-K-(QZ-(Yn2-P)m2)j wherein P is a phosphonic group or a phosphonic salt, as for example, a phosphoric group substituted with one or more alkali metals;
Y is -OCO-, -NRiCO-, or -CON(Rl)R2-;
R~ is H, CH2C02H, or substituted or unsubstituted alkyl;
R2 is substituted or unsubstituted alkyl, aryl, or arylalkyl;
Q1 and Q2 are substituted or unsubstituted aryl groups;
K is H, -NH-CO-NH-, -NH-CS-NH-, -NHCO-R3-CONH-, or -NHCS-R3-CSNH-; provided that when K is H, j is 0;
R3 is a substituted or unsubstituted aryl group;
j is 0, 1, or 2;
nl and n2 are independently 0, 1, or 2; and ml and m2 are independently an integer from 1 to 4.
Embodiments of the phosphoric acid agents of the present invention comprise compounds of the formulae:
A) CP_Yn1 \ N.B~N \ Ynz P~m2 or B) Yn~-P) ~ m~

wherein Y, P, nl, n2, mi, m2 R3 are as defined above;
B is CO, CS, CO-R3-CO, or CS-R3-CS;
SUBSTITUTE SHEET (RULE 26) R4 and RS are independently H or a substituted or unsubstituted alkyl group;
R6 is H, or NCOR'; and R' is aryl, substituted aryl, or nitro substituted aryl.
Preferred phosphoric acid agents of the invention are set forth below in Tables 1-3, 5 which tables provide chemical formulae, molecular weights, and properties of the compounds including some embryo and inhibition data.
Synthesis of Phosphoric Acid Agents The syntheses of the phosphoric acid agents of this invention are shown in process Schemes 1 and 2 below. General procedures for the syntheses are described under Synthetic Methods A-G. Synthetic methods used for the synthesis of the individual phosphoric acid agents are listed in Tables 2-3.
The phosphoric acid agents synthesized herein can be divided into three general groups. Table 1 shows phosphoric acid agents with small urea bridges. The basic structural formula, the chemical substitutions at each position, formula and molecular weight, are shown for each compound. The molecular weights of this group varied from 416 to 614 depending on the substitutions at positions 2, 3 and 4 of the basic structure. The basic structure of this group was synthesized using synthetic methods B and C described below (see structure for NF
158 and NF 161). Additional substitutions at positions 2, 3 and 4 were made using synthetic methods A, Fa, Fb and G as indicated in Table 1.
Table 2 shows seven phosphoric acid agents with large urea bridges. Two basic structural formulas with four benzene rings are shown with the chemical substitutions at each position, formula and molecular weights for each compound. The molecular weights of this chemical group varied from 698 to 855 depending on the substitutions at 2, 3, 4, 4' and X of the basic structures. The basic structures of this group were synthesized using synthetic methods A, B, C and D indicated below. (See NF 067, 068, 069 and 681). NF 540 and NF
544 required synthetic steps Ea and Ec instead of D.
Table 3 shows four phosphoric acid agents with miscellaneous structures.
Because of differences in the basic structures of this group, the entire structure is shown with the formula and molecular weights. The molecular weights of this group varied from 424 to 949. NF 166 was synthesized using synthetic method A described below. NF I67 required synthetic method A, B and C, whereas NF 050 and NF 542 required synthetic method A, B, A, B and C.
SUBSTITUTE SHEET (RULE 26) WO 99/05148 PCTIUS9$/15470 ~r b M

N

O

w O o O V1 N M ~O
r-~.

M ...O N d' -m ~t N N N N N N
W

G

O

'.- e~

O

M ~ ~

o O O U

_ ~ ~ ~ O o o ~

N O x .~ x x x x Q
tL~ , O

' U ' O z =-z ~ x ~

O ' O ~ J

=_z . z w o z ~ ,.~, U
O U ~

~M x x x z x a x a o Z
U

a ~''~ '~ O

U

O

N x x o x x x x x x x .

3 d ~

~ w 'a w w U U

i ~ , c as o U U U U ~ ~ U o w w 1 o ~ U U tn a0 as CG fz :o ~ ~

v~ a1m t7 C7 V C7 w w Q
~

N N N N N N N N M M

~ ~ ~ ~ a O O O O M O O c vw n ~n ~n ~1v, vG vo n d1 ~

CSI~ CASCd z z z z z z z z z z x x x x x ~ x x x x w U U

U U U U U U U U

o H

x M

O ~ v0 h N 01 h N N M ~t i .-..r ~' !!'M ~' ~Tt1'd' ,.~ ., 'O ~Zz z z ~Z ~Z z ~Z ~Z z H v w w SUBSTITUTE SHEET (RULE 26) eri N
O
O
,~.r ~ ~ o ~ O
w ca ~t W N N 3t W N N
O
x x ~r x x v v z N
I ~ N ~ ~ O CIA I ~ N z O O
_-_ _-_ "~'~'' ~'~e~ Gn O
rv ~ ~ ~ rv =~ x ° U U
- .°- z z - o _-_ ~ x x =-_ o ~~ ~~ ~O O
_-_ ~ o 0 x ~. ~. _-_ _ a ~ M ° x x x ~I o ~I o N
...
O O
v~ V Z = w. _ SIN ~ SIN ~N x x x v Mo. x x v v w c ai as a a A A A ~ A A c~ xi ~~ 'b U U U ~~ 'd U U v ci ~ ~ fn GA f~ ~ ~ ~'p ~q W W
-c ~ a ~~Q d a ~~ a a w w ~3 wao o ~~~ ~~~o~o c~
~ e0a c~ ~ Z ~a a s ed z z ~ z z z b.°c ~ ~ ~ aQ ~ ~ w O O O O O O O
z z z z z z z O x x x x x x x > w U U U U U U U
a~
o ~ .... N oo c~ o ,= >C .fl o vc o o v't', v., v z z ~Z ~Z z z z suesn~ure sHesr ~u~ Zs~
Table 3. The code number, formula, molecular weight, synthesis method, and chemical structures of phosphonic acid agents are indicated.
Synthesis Method Compound A NaH03P
~N ~ H
H \ I N~P~Na O
NF 166 C~2Ht~N20gP2Na2 (424.19) NOy \ I
A,B,C

H ~ H
N \ I N
I
NaH03P \ O O w P03HNa NF 167 C27H2oN40, lP2Na2 (684.40) H ~ O H H O \ H
N a2p~P / N \ I N ~ H N \ N I i N ~ P03N ay A, B, A, if \ 0 H \ O / H O \
B, C
NF 050 C4,H3oN60aPzNa4 (936.62) H H

A~ B~ A~ Nw I w i Po~HN~
B C i I O H H O I \
\ N , N N w N
H3 H \ I ~ I i Fi H3 NF 542 C4sHaoN64~ ~PzNax (948.77) SUBSTITUTE SHEET (RULE 26) *rB
Schemes 1 and 2 as set forth below show general procedures for preparation of the novel compounds of this invention. In Scheme l, the phosphoric acid agents are prepared by initial reduction of a vitro benzene phosphoric acid to the amino derivative. This amino derivative is then reacted with a di-acid halide to yield the phosphoric acid substituted agent (e.g. compounds 4a, b). Such di-acid halides include phosgene, thiophosgene and a dicarboxylic acid halide substituted aryl group.
In an alternative embodiment, the amino derivative is reacted with a vitro benzoyl halide in a buffered medium and at temperatures of from about 20 to about 40°C. The nitrobenzoyl halide is added in an organic solvent such as toluene. At the conclusion of the reaction, the aqueous layer and organic layer are separated and the aqueous layer is acidified by the addition of a mineral acid from which a precipitated intermediate product, nitrobenzamido-benzenephosphonic acid, is recovered.
This nitrobenzamido-benzenephosphonic acid is then hydrogenated in the presence of a hydrogenation catalyst comprising a precious metal such as palladium or platinum on carbon to hydrogenate the vitro group and form an aminobenzamido-benzenephosphonic acid.
The aminobenzamido-ben~enephosphonic acid is then dissolved in a buffered aqueous medium and treated with phosgene in an organic solvent to produce the final agent precipitated product. These reactions are described more specifically under Procedures A, B
and C.
Scheme 2 sets forth a general synthesis for producing the various arylalkyl-, phenoxycarbonyl-, and carbamoyl-phosphoric acid analogues. In these reactions, the substituted benzene derivatives, compounds 11, 15 or 17 are reacted with the appropriate alkyl phosphite to form compounds 13, 16 and 18, respectively. These compounds will contain the appropriate substituents as shown in Scheme 2.
More specifically, the chloromethyl substituted benzene 12 is reacted with an alkyl phosphite by heating at about 80°C up to reflux for 3 to 10 hours. The resulting 12 is then nitrated to 13. Compound 14 may then be produced from compound 13 by heating with concentrated hydrochloric acid and crystallizing the product.
SUBSTITUTE SHEET (RULE 26) Scheme 1 General synthesis of the phosphoric acid substituted agents:

H~ 1I ~ NH2 + CI 1I ~ N~ ~ H2~ 1 ~ N O
/ 4 4. I / ~ I NOp 3 3, ~H~
3' 1a,b 2a,b 3cf H~ t o H2~' t W .B / 1 ~aH2 I W N 1 I N ~N I / 4 H I / NHZ
~H H~ 3 3 g 3' 5c-f 4a, b H
H~ I ~ H I N o N I H \ I ~3 2 H,~H
sc f o s o 0 s: ~ J~ / i R
a 1, 3 b 1, 4 c 1, 3 1', 3' d 1, 3 1', 4' a 1, 4 1', 3' f 1, 4 1', 4 SUBSTfTUTE SHEET (RULE 26) Scheme 2 General synthesis of arylalkyl-, phenoxycarbonyl- carbamoyi-phosphoric acid agents 0'P (OCH312 O~~P,(~3Yt p'P (OHYt CI
R / P(OCH3y~R R R
/ HN03lHyS04 / H20/HCI /
-~ ~ ~ - a ~

11a R=H 12a R=H 13a R=H 14a R=H

11b R = CHZCI12b R = CFi2P0(OCH3~13b R = CHyPO(OCH3~14b R =
CHpP03H2 O
P(~~3I3 ~ / (1 O~~P~~H3 '~ 3 4~ O 4~ O
O
ISa~C 18a-c ~) CI-SKCHS)3 / ~ X~P~~ a 2) H20, NaOH ~ 4 ~
H 0 198-0 X = O
~''N=C=O ~ N p-~H3 20a, b X = NH
O~i4'r~\~~ HOP(OCH~ ~ / I ~ ~~H3 ---~ ~ O
17a, b 18a, b a: 4-N02 b: 3-N02 c: 2-N02 SUBSTITUTE SHEET (RULE 26j In a further embodiment, the nitrophenoxy carbonylchloride 15 is reacted with an alkylphosphite in an exothermic reaction to produce a nitrophenoxycarbonyl-phosphonic acid dialkyl ester 16. This intermediate can be converted by reaction with sodium iodide in a solvent with an haloalkylsilane by heating at about 30° to about 50°C to produce compound 19.
Alternatively, the nitrophenylisocyanate (compound 17) is reacted with a dialkylphosphite in an exothermic reaction to yield the phosphonic acid ester (compound 18).
Thereafter, the phosphonic acid ester is reacted with trisodium iodide in a solvent and with an halotrialkylsilane by heating at about 30° to about 60°C to produce compound 19.
Compound 16 can be converted to compound 19 by the same procedure.
A. 4-(4-Nitrobenzamido)-benzenephosphonic acid (3f) 4-Aminobenzenephosphonic acid (lb) was prepared as described by Doak D.O. and Freedman CD, J. Amer. Chem. Soc., 74, 753 (1952). Compound lb_(17.3 g, 0.1 mot) was dissolved in 2 N NaOH (100 ml). To this solution (pH 8), a solution of 4-nitrobenzoylchloride (2b, 30 g, 0.15 mot) in toluene (100 ml) was added dropwise at room temperature with vigorous stirring. Stirring was continued until no more lb was detectable by TLC. Soon after the beginning of the addition of 4-nitrobenzoylchloride, 3f began to precipitate.
NaOH (2 N) was added to the reaction mixture at a rate that kept 3f in solution. At the end of the reaction, the aqueous layer was pH 11. The toluene layer was separated. The aqueous layer was acidified to pH 1.0 by addition of concentrated hydrochloric acid and stirred for 30 min.
The precipitate was filtrated and crystallized from methanol.
Yield 25.3 g (78%). Melting point: 257-260°C.
C~3H"N2O6P (322.4) In a similar way a series of aminoaryl- or aminoalkyl-phosphonic acids was benzoylated using nitrobenzene-mono or di-carbonylchlorides.
B. 4-(4-Aminobenzamido~benzenephosphonic acid (Sf) Compound 3f (8.05 g) was dissolved in a mixture of 1 M Na2CO3 (15 ml), water (55 ml) and methanol (30 ml) (pH 8.0) and hydrogenated at room temperature at normal pressure using SUBSTITUTE SHEET (RULE 26) 10% Pd/C (200 mg) as catalyst. After absorption of the theoretical amount of hydrogen (about 4 hr), the filtrate of the reaction mixture was concentrated to 70 ml and acidified with 2 N HCl to pH 6Ø The monosodium salt of compound 5f precipitated. It was washed with water and dried at about 40°C. C~3H~3N2OaP (292.4); C,3H~2N20aPNa (314.4) monosodium salt of compound S 5f.
In a similar way all aromatic vitro compounds used as intermediates were hydrogenated to the corresponding aromatic amino compounds.
C. 4,4'-[Carbonylbis(imino-4,1-phenylencarbonylimino)J-bis-benzenephosphonic acid (6f).
Compound 5f (5.85 g, 20) was dissolved in water (50 ml) and 5 M NaOH (4.0 ml).
To this solution, a 20% solution (2 M, 50 ml, 100 nmol) of phosgene in toluene was added dropwise (4 hr) under vigorous stirring. During the whole reaction time, the pH of the reaction mixture was maintained at pH 5.0 by automatic addition of 5 M NaOH. A suspension was formed during the addition of phosgene. The toluene layer was separated. The aqueous suspension was acidified with 2 N HCl to pH 1 and stirred for 30 min. The precipitate was filtered by suction, stirred three times with water (20 ml, 30 min) and dried.
Yield: 3.6 g (61%) C27H24N409P2 (610.4) In a similar way amino derivatives were reacted with a di-acid halide to yield the phosphoric acid substituted agent. A thiophosgene or a dicarboxylic acid halide was used to synthesize the corresponding agent.
D. Tetrasodium salt of compound 6f Compound 3f (0.61 g 1.0 mol) was suspended in water (10 ml). The suspension was titrated very slowly with 0.1 N NaOH to pH 9Ø The resulting clear solution was evaporated i.
vacuum to dryness.
CZ~HzoNa09P2Na4 (698.4) TLC (Acetonitrile, NH3 cone, water 6+2+2); compound 6f Rf 0.63; compound Sf ltf 0.75;
compound 3f ltf 0.79; 4-Nitrobenzoic acid ltf 0.75; HPLC purity of compound 6f > 96%
determined by the method described by Kassack and Nickel (1996).
SUBSTITUTE SHEET (RULE 26) E. 4-Nitro-1,3-xylene-a,a-diphosphonic acid (14b) a) 1,3-Bis-(chloromethyl)benzene (llb, 25 g, 0.1 mol) and triethyl phosphite (36.5 g, 0.22 mol) were heated under reflux for 10 hr.
b) The reaction mixture (12b, yellow oil) was cooled to room temperature and added dropwise to a mixture of conc. nitric acid (30 ml, 0.4 mol) and conc. sulfuric acid (30 ml) at about 0-5°C. After 2 hr, the reaction mixture was poured in ice water (500 ml) and extracted with toluene. The toluene extracts were washed with water and evaporated, yielding a yellow oil (13b).
c) Compound 13b was heated with conc. hydrochloric acid under reflux for 20 hr. The reaction mixture was evaporated. The residue was crystallized from water yielding yellow crystals of compound 14b (mp 172°C, 15.8 g, yield = 50%).
In a similar way using steps a) and c) (4-nitrophenyl)methane phosphoric acid (I4a) was synthesized starting from 4-vitro-a-chlorotoluene.
F. (3-Nitrophenyl)carbamoyl phosphoric acid monosodium salt (20b).
a) 3-Nitrophenylisocyanate ((17b) 0.82 g, 5 mmol) and dimethyI phosphite (0.55 g, 5 mmol) were mixed. Addition of two drops of triethylamine led to a spontaneous exothermic reaction. The reaction mixture was crystallized from methanol/ether yielding yellow crystals (0.44 g, 32%), mp 107°C. (3-nitrophenylcarbamoyl phosphoric acid dimethylester,18b).
b) Compound 18b (5 mmol) and sodium iodide (10 mmol) were dissolved in acetonitrile (30 ml). Chlorotrimethylsilane ( 15 mmol) was added dropwise. The mixture was heated to about 40°C for 30 min, cooled and filtrated. The filtrate was concentrated to a volume of 10 ml. Water (10 ml) was added and the pH of the solution was adjusted to pH 3 by addition of 0.1 N NaOH.
The solution was extracted three times with ether ( 15 ml). The aqueous phase was freeze-dried, yielding an amorphous powder (20b).
G. 4-Nitrophenoxycarbonylphosphonic acid dimethylester (16a). Trimethyl phosphite (4 ml, mmol) was added dropwise under stirring and cooling to 4-nitrophenoxycarbonyl chloride SUBSTITUTE SHEET (RULE 26) (15a, 6.4 g, 32 mmol) (exothermic reaction). The reaction mixture was crystallized from ether/pentane yielding yellow crystals (16a, 6.8 g, 77%), mp 105°C.
In a similar way the 2-nitro- (16c) and 3-nitro- (16b) analogues were synthesized. (See also Doak GO and Freedman CD. J. Amer. Chem. Soc., 74:753, 1952. Kassack M and Nickel P.
5 J. Chromatogr. B, 686, 275-284, 1996, incorporated herein by reference in their entirety).
The basic structure of the molecules synthesized were similar to suramin.
However, the trisulfonic acid derivatives were replaced with phosphoric acid groups (P03HNa) or (-O-CO-P03HNa) at the 2 (2'), 3 (3') or 4 (4') positions. These alterations resulted in lower molecular weights (414-950) compared to the molecular weight of suramin (1429) and other 10 trisulfonic acid analogues (1162-1541). Both large urea and small urea phosphoric agents were synthesized. The chemical and pharmacological properties are very different from those described for other suramin analogues. The metabolic clearance rates of the phosphoric acid agents appear to be much faster (hours versus 45-55 days for suramin).
Pharraacological Activities: Antiangiogenic Activity in the Chick Chorioallantoic Membrane Assay In Vivo and the Human Microvascular Endothelial Cells In Vitro.
The purpose of this experiment was to test the ability of the phosphoric acid agents described above to inhibit angiogenesis. The ID50, the dose that produces 50%
inhibition of angiogenesis, was determined for suramin and each of the phosphoric acid agents by measuring the ability of various doses to inhibit angiogenesis in vivo in the chick egg chorioallantoic membrane (CAM) assay as described by Gagliardi et al. 1992. Some of the phosphoric acid agents showed ID50 values significantly lower than suramin. Two phosphoric acid agents, NF
069 and NF 681, showed the lowest ID50 values (4-8 times more active than suramin). Six other phosphoric acid agents showed antiangiogenic activity that was 2 to 4 times greater than suramin (NF 050, NF 067, NF 161, NF 167 and NF 428). Other phosphoric acid agents were equipotent to suramin. These data clearly indicate the discovery of a group of unique phosphoric acid agents which show antiangiogenic activity that is up to 8 fold greater than suramin in the CAM assay.
It was also found that the compounds described above, in general, showed a closely related antiangiogenic activity on the growth of human dermal Microvascular endothelial cells SU6STiTUTE SHEET (RULE 26) WO 99/05148 PCT/US981154?0 stimulated by basic fibroblast growth factor (bFGF) in vitro. The IC50 values for those phosphoric acid agents were 22-80 pM compared to 438 pM for suramin. This indicates that these phosphoric acid agents are 5-40 times more potent inhibitors of endothelial cell growth than suramin in vitro.
Differences in the Effect of Suramin and the Phosphoric Acid Agents on Angiogenesis in the 6-Day Old and 11-Day Old CAM Assay.
According to Ausprunk et al. (1974), capillary angiogenesis in the CAM is completed by day 11. Measurements of intercapillary distances are also consistent with the cessation of capillary growth after day 10. Flamme et al. ( 1991 ) showed that CAM fluid contains angiogenic growth factors, that the mitogenic activity of these growth factors was temporally related to the vascular growth in the CAM, and that by day 10, there was a sharp decrease in growth factor activity in the CAM fluid which preceded the termination of capillary growth by one day. Based on these observations, the effect of suramin and some phosphoric acid agents on the established vessels of the CAM membrane after cessation of vascular growth was determined.
The implants were prepared as previously described by Gagliardi et al. (1992), implanted on day 11 and read on day 13.
The results of this study showed that the phosphoric acid agents (NF 067, NF
069, NF
681 ) were very potent inhibitors of angiogenesis in the 6-day CAM and exhibited no significant inhibitory activity in the 11 day CAM (see Table 4). Suramin showed antiangiogenic activity in both the 6-day and 11-day CAMS.
This is a very important finding and suggests that the phosphoric acid agents are very potent inhibitors of angiogenesis during the phase of rapidly growing vessels in the CAM (6-day) but do not affect the established vasculature (11-day CAM). On the other hand, suramin clearly affected both the growing and established vascular cells. These results indicate that the mechanisms of action of suramin and the phosphoric acid agents on angiogenesis are different.
Additional experiments compare the mechanisms of actions of the phosphoric acid agents on angiogenesis in vivo.
SUBSTITUTE SHEET (RUL.E 26) Table 4. Comparison of the inhibition of angiogenesis by 70 nmol of suramin or a equimolar amount of a phosphoric acid agents in the 6-day and 11-day chick egg chorioallartoic (CAM) assay.

Chemical 6-day CAM 11-day CAM

Analogues Structure % Inhibition % Inhibition suramin 64 50 NF 068 phosphoric 0 0 NF 067 phosphoric 63 0 NF 069 phosphoric 68 0 NF 681 phosphoric 100 0 Differences in the Inhibitory Effects of Suramin and the Phosphoric Acid Agents on bFGF-Stimulated and Non-Stimulated Endothelial Cells.
Suramin is a highly charged molecule with six sulfonate groups that are ionized at physiologic pH. This results in significant nonspecific binding to polypeptide growth factors (Coffey et al., 1987). However, suramin also exhibits specificity by binding to specific sites on a growth factor, similar to heparin binding to bFGF (Middaugh et al., 1992).
While these studies showed that suramin is able to disrupt the binding of growth factors to their receptors in intact cells, it also has diverse effects on other key enzymes involved in signal transduction and mitogenesis that probably contribute to its antiproliferative and antimetastatic activities.
The data indicates that bFGF induced a dose-related increase in total protein and total DNA in human dermal microvascular and porcine pulmonary artery endothelial cells. The presence of suramin (210 uM) inhibited the stimulatory effect of bFGF, as has also been reported by Takano et al. (1994) and Braddock et al. (1994) for bovine aorta and bovine adrenal microvascular endothelial cells. In the presence of much lower concentrations (25 pM), the phosphoric agents, NF 050, NF 067, NF 069, NF 681, NF 161, NF 167 and NF 428, not only inhibited the stimulation of endothelial cell growth by bFGF but significantly reduced total protein content far below the unstimulated control endothelial cells. These exciting results show that the agents are much more potent inhibitors of endothelial cell growth than suramin and that SUBSTrtIITE SHEET (RULE 26) mechanisms other than blocking growth factor binding to endothelial cells play a very important role in their antiangiogenic activity.
The results with suramin are similar to those reported in the literature (Takano et al., 1994 and Braddock et al., 1994) that suramin inhibited Ilzs bFGF binding in a dose-related manner.
However, the phosphoric agents in the same molar concentrations used for suramin did not affect either the total or the specific binding of the iodinated growth factor to the endothelial cells. The percentage of inhibition of I125 bFGF binding to low and high affinity binding sites of human microvascular endothelial cells was 96% at 70 uM of suramin, decreasing to 9%
at 25 1cM of suramin. However, for the same concentrations of the phosphoric acid agents, the inhibition of growth factor binding was always less than 5% in relation to the control.
These data clearly show that we have identified a chemical group that are very potent as antiangiogenic agents and express their activity through different mechanisms than those widely accepted for suramin.
As described above, the phosphoric acid agents are very inhibitory in the actively growing vessels of the 6-day CAM but showed almost no activity on the established vessels of the 11-day CAM (see Table 2). When compared with the MTT assay confluent and low density cultures in log phase growth, the phosphoric acid agents are 10-fold more inhibitory for growing cultures than for confluent human microvascular endothelial cell cultures.
This in vitro finding corroborates our data with the CAM assay in different phases of growth, suggesting that this new class of phosphoric acid agents target growing blood vessels.
Differences in the Growth Inhibitory Effects of Suramin and the Phosphoric Acid Agents in Human Microvascular Endothelial Cells and Established Cancer Cell Lines in Culture.
We have investigated the activity of the phosphoric acid agents on some tumor cell lines proliferation in vitro and compared this activity with the antiangiogenic activity of these compounds in the CAM assay and their inhibitory activity in human microvascular endothelial cell growth.
The MTT assay (Carmichael et al., 1987) was used to examine the effects of suramin and selected phosphoric acid agents on cell proliferation. Suramin and the phosphoric acid agents inhibited cell proliferation in a dose-related manner. Analysis of the inhibitory action of the SU9STtTUTE SHEET (RULE 26) phosphoric acid agents in adrenal cortex carcinoma (SW13), human pancreatic adenocarcinoma (CFPAK-1), human prostate carcinomas (LNCap and PC3) showed IC50 equipotent or values less than suramin. In other cell lines, such as human breast carcinomas (MCF7 and T47D), human fibrosarcoma (HT1080), human colon adenocarcinoma (CaCo2), human glioblastoma (LJ87), kidney carcinoma (A498) and lung carcinoma (A427), the phosphoric acid agents showed IC50 values higher than suramin.
The phosphoric acid agents, NF 067 (which is 20 times more potent than suramin in inhibiting microvascular endothelial cell growth), expressed very low antiproliferative activity against different cancer cell lines in vitro. Our data and the reports in the literature strengthen our important finding that some of the phosphoric acid agents are more potent inhibitors of angiogenesis in the CAM assay and to human microvascular endothelial cell growth than suramin. This effect is not observed with some cancer cell lines. This suggests that there is specificity for endothelial cells in the inhibitory effect of the phosphoric acid agents not observed with suramin and the other trisulfonic acid analogues.
The toxicity of suramin and the phosphoric acid agents in vivo in mice.
A limitation on the clinical use of suramin is the narrow margin between the dose required to achieve anti-tumor activity and that leading to the onset of prohibitive toxic side effects.
Suramin toxicity has been reviewed by LaRocca et al. ( 1990). It is clear that compounds with similar antitumor activity to suramin but with substantially lower toxicity would be of considerable potential therapeutic value as an antitumorigenic or antiangiogenic agent. Toxicity studies were performed with suramin, three sulfonic analogues more potent (2 times) than suramin in relation to inhibition of angiogenesis and endothelial cell growth and four phosphoric agents (10 to 40 times more potent). Mice were injected intraperitoneally with 0-150 ~M/kg body weight of the compounds to be tested, every other day for a total of five injections. The animals were carefully observed daily and weighed every third day for 28 days after the last injection. After the 28-day observation period, the animals were euthanized, blood was collected through cardiac puncture and the following tissues were subjected to histological investigation:
heart, lungs, liver, spleen, adrenal gland, kidney, sciatic nerve, soleus muscle and brain.
SUBSTITUTE SHEET (RULE 26j Animals treated with suramin at the highest dose ( 150 pM/kg body weight) died before completion of the five injections. We observed poor coat condition, weight loss, eye irritation and lacrimation by the end of the five injections in animals treated with suramin. The poor coat condition and reduction of 10-15% in body weight occurred at 150, 75 and 35 uM/kg body 5 weight during the injection period. The coats and eye irntation became better but not nornial and the body weight stabilized but did not return to normal over the subsequent 28 days of observation. A similar response was seen in two of the three sulfonic acid analogues that were tested. On the other hand, no mice treated with the four phosphoric acid agents died during the acute injection phase. Furthermore, their body weight did not decrease but they continued to gain 10 weight at the same rate as the control animals at all levels of treatment.
The body coat was normal and no eye irritation was noted in all animals treated with phosphoric acid agents.
Histological analysis of the tissues from animals treated with suramin showed a dose-related frequency of lipoid degeneration of the zona reticularis of the adrenal gland and vacuolar changes in the proximal convoluted tubules of the renal tubular epithelium. The animals 15 treated with equimolar doses of the phosphoric analogues showed no significant changes in any important pathologic microscopic findings in those tissue samples.
These studies clearly indicate that the phosphoric acid agents show significantly lower toxicity than suramin and its trisulfonic acid analogs.
20 EXA~lELES
Structure-activity relationship for the antiangiogenic activity of the phosphoric acid agents growing vessels in the chick chorioallantoic membrane assay.
The phosphoric acid agents are extremely potent antiangiogenic compounds with molecular weights that are about half that of suramin. The phosphoric acid agents are up to 30 times more active than suramin in the CAM assay. Furthermore, the nonspecific binding of these compounds to serum proteins is lower than suramin and probably, as a consequence, a higher proportion is available in the free form to the cells and the half life is shorter. The size of the molecule also appeared important. Ten of the phosphoric acid agents have molecular weights of less than 600 and contained small central urea bridges (see Table 1), showed less antiangiogenic activity than the seven phosphoric acid agents which have big central urea bridges and higher SUBSTITUTE SHEET (RULE 26) molecular weights (650-900) (see Table 2). A third group of four phosphoric acid agents with miscellaneous structures is shown in Table 3.
There is considerable variation in antiangiogenic activity in compounds of similar structure. For example, NF 068, which is structurally very similar to NF 067, NF 069 and NF
681 (all are phosphoric acid agents with the same molecular weight), showed a substantial difference in the antiangiogenic activity (0% inhibition) in comparison with the same concentration of NF 069 and NF 681 (90% inhibition). This suggests that slight steric modifications in the molecule can induce dramatic changes in the potency of inhibition of angiogenesis, opening new avenues for antiangiogenic drugs design.
The structure activity relationship for Inhibition of bFGF induced cell growth in human microvascular endothelial cells.
Dose response curves were determined for the most active agents in human microvascular endothelial cell cultures treated with various concentrations in the presence or absence of bFGF.
Inhibition of cell growth was evaluated by measuring total protein and total DNA and the IC50 was estimated for each dose response curve. The most potent compounds in relation to the inhibition of bFGF-induced endothelial cell growth were the phosphoric acid agents.
Our results show a close correlation between the inhibition of angiogenesis in vivo and the inhibition of endothelial cell stimulated growth in culture for the selected agents.
Among the five most active agents in vitro, four were phosphoric acid agents, the same compounds which were the most antiangiogenic in the CAM assay. These results suggest that the inhibition of angiogenesis in vivo in the CAM assay may involve a specific suppression of endothelial cell growth. Thus, we have identified a specific group of phosphoric acid agents with significantly lower molecular weight, less charged and much higher antiangiogenic activity than suramin and its sulfonic acid analogs.
The finding that a unique chemical group of the phosphoric acid agents, are much more potent as antiangiogenic compounds, less toxic and with shorter half fifes than suramin, suggest that the phosphoric acid agents are of clinical significance and widen the therapeutic window for treatment of angiogenic dependent diseases.
SUBSTITUTE SHEET (RULE 26) Differences in the inhibitory effects of suramin and the phosphonic acid agents to bFGF-stimulated and non-stimulated endothelial cells. Suramin is a highly charged molecule with six sulfonate groups that are ionized at physiologic pH. This results in significant nonspecific binding to polypeptide growth factors (Coffey et al., 1987).
However, suramin also exhibits a degree of specificity by binding to specific sites on a growth factor, similar to heparin binding to bFGF (Middaugh et al., 1992). While these studies showed that suramin is able to disrupt the binding of growth factors to their receptors in intact cells, it has diverse effects on other key enzymes involved in signal transduction and mitogenesis that probably contribute to its antiproliferative and antimetastatic activities. Our data showed that bFGF
induced a dose-related increase in total protein and total DNA in human dermal microvascular and porcine pulmonary artery endothelial cells.
The presence of suramin (210 pM) inhibited the growth factor stimulatory effect, as has also been reported by Takano et al. ( 1992) and Braddock et al. ( 1994) for bovine aorta and bovine adrenal microvascular endothelial cells. In the presence of much lower concentrations (25 pM), the phosphonic agents, NF 067, NF 069 and NF 681, not only inhibited the stimulation of endothelial cell growth by bFGF but significantly reduced total protein content far below the unstimuiated control endothelial cells.
These results show that the phosphonic acid agents are much more potent than suramin in inhibiting angiogenesis and that mechanisms other than blocking growth factor binding to endothelial cells play a very important role in their antiangiogenic activity.
Results with suramin are similar to those reported in the literature (Takano et al., 1992 and Braddock et al., 1994) that suramin inhibited bFGF binding in a dose-related manner; but the phosphonic analogues in the same molar concentrations used for suramin did not affect either the total or the specific binding of the iodinated growth factor to the endothelial cells.
The percentage of inhibition of h2s bFGF binding to low and high affinity binding sites of human microvascular endothelial cells was 96% at 70 pM of suramin, decreasing to 9% at 25 gM
of suramin. However, for the same concentrations of phosphonic acid agents, the inhibition of gmwth factor binding was always less than 5% in relation to the control. These data clearly show SUBSTITUTE SHEET (RULE 28) that there is identified a chemical goup of phosphoric acid agents that are very potent as antiangiogenic agents that express their activity through different mechanisms than those widely accepted for suramin.
The phosphoric acid agents are very inhibitory in the actively Bowing vessels of the S 6-day CAM but showed almost no activity on the established vessels of the 11-day CAM. When compared with the MTT assay confluent and low density cultures in log phase gowth, the phosphoric acid agents are 10-fold more inhibitory for Bowing cultures than for confluent human microvascular endothelial cell cultures. This in vitro finding corroborates our data with the CAM
assay in different phases of gowth, suggesting that this new class of suramin analogues target Bowing blood vessels and does not effect established blood vessels.
Differences in the growth inhibitory effects of suramin and the phosphoric acid analogues in human microvascular endothelial cells and established cancer cell lines in culture.
1 S We have compared the activity of suramin and the phosphoric acid agents on some tumor cell lines proliferation in vitro and compared this activity with the antiangiogenic activity of these compounds in the CAM assay and their inhibitory activity in human microvascular endothelial cell gowth. The MTT assay (Carmichael et al., 1987) was used to examine the effects of suramin and selected phosphoric acid analogues on cell proliferation. Suramin and the phosphoric acid agents inhibited cell proliferation in a dose-related manner.
Analysis of the inhibitory action of suramin and the phosphoric acid agents in adrenal cortex carcinoma (SW13), human pancreatic adenocarcinoma (CFPAK-1), human prostate carcinomas (LNCap and PC3) showed IC50 value equipotent or less than suramin. In other cell lines, such as human breast carcinomas (MCF7 and T47D), human fibrosarcoma (HT1080), human colon adenocarcinoma (CaCo2), human gliobiastoma (U87), kidney carcinoma (A498) and lung carcinoma (A427), the phosphoric acid agents showed equipotent or lower inhibitory activity in comparison to suramin.
Our data strengthened our important finding that the phosphoric acid agents are much more potent inhibitors of angiogenesis in the CAM assay and to human microvascular endothelial cell gowth than suramin. This effect is not observed with some cancer cell lines. This suggests SUBSTITUTE SHEET (RULE 26) that there is specificity for endothelial cells in the antiangiogenic effect of the phosphoric acid agents.
Comparative Toxicity of suramin, trisulfonic analogues and the phosphoric acid agents in vivo in mice.
A limitation on the clinical use of suramin is the narrow margin between the dose required to achieve anti-tumor activity and that leading to the onset of prohibitive toxic side effects.
Suramin toxicity has been reviewed by LaRocca et al. (1990). It is clear from studies so far that compounds with equipotent or greater antitumor activity but with substantially lower toxicity are of considerable potential therapeutic value as an antitumorigenic or antiangiogenic agent.
Preliminary toxicity studies were performed with suramin, three sulfonic analogues more potent (2 times) than suramin in relation to inhibition of angiogenesis and endothelial cell growth and the four most active phosphoric analogues (up to 30 times). Mice were injected intraperitoneally with suramin or equimolar doses of the sulfonated analogues or the phosphoric analogues or the phosphoric acid agents (1-150pM/Kg body weight) every other day for a total of five injections.
The animals were carefully observed daily and weighed every third day for 28 days after the last injection. After the 28-day observation period, the animals were euthanized, blood was collected through cardiac puncture and the following tissues were kept for histological investigation: heart, lungs, liver, spleen, adrenal gland, kidney, sciatic nerve, soleus muscle and brain.
Animals treated with suramin at the highest dose ( 150 pM/Kg body weight) died before completion of the five injections. Poor coat condition, weight loss, eye irritation and lacrimation was observed by the end of the five injections in animals treated with suramin. The poor coat condition and reduction of 10-15% in body weight occuried at 150, 75 and 35 pM/Kg body weight during the injection period. The coat condition and eye irritation became better but not normal and the body weight stabilized but did not return to normal over the subsequent 28 days of observation. On the other hand, mice treated with the phosphoric acid agents did not die during the acute injection phase. Furthermore, their body weight did not decrease but they continued to gain weight at the same rate as the control animals at all levels of treatment. The body coat was normal and no eye irritation was noted.
SU6STfTUTE SHEET (RULE 26) Histological analysis of the tissues from animals treated with suramin showed a dose-related frequency of lipoid degeneration of the zona reticularis of the adrenal gland and vacuolar changes in the proximal convoluted tubules of the renal tubular epithelium. The animals treated with equimolar doses of the phosphoric acid agents showed no significant changes in any S important pathologic microscopic findings in those tissue samples. These preliminary studies clearly indicate that the phosphoric acid agents show a greater than 10-fold increase in antiangiogenic activity and significantly lower toxicity.
Structure of Phosphoric Acid Agents The phosphoric acid agents studied were selected based on the criteria of drug potency in relation to inhibition of angiogenesis in the CAM and inhibition of microvascular endothelial cell growth in vitro, availability, chemical purity and sampling of each different chemical structure subgroup. Twenty-two phosphoric acid agents, including NF 067, NF 068 NF 069, NF 681, and 1S NF 162, synthesized by our laboratory, are used in various concentrations.
In the CAM assay, the m50 for suramin was 7S nmol and the IDSO for the phosphoric acid agents, NF
069, NF 681 and NF 067, was respectively, 9, 2 and 32 nmol. The estimated ICSO for suramin in the bFGF-stimulated human microvascular endothelial cells was 437 pM and for NF
069, NF 681 and NF 067 were respectively, 7S, 1.S and 19.4 pM, reflecting activity that is up to 200 times more potent than suramin. NF 068 is a closely related compound chemically which does not show any antiangiogenic activity. The structures of these phosphoric acid agents and suramin are shown in Tables 1-3.
The inventors have identified a clear correlation between the chemical structure and antiangiogenic activity. The phosphoric acid agents are far more potent inhibitors of 2S angiogenesis and bFGF-stimulated endothelial cell growth than any suramin.
Furthermore, the phosphoric acid analogues with large central urea bridges are in general more active than the group with small central bridges or other configurations.
SUBSTITUTE SHEET (RULE 26) Effects of the phosphoric acid agents and suramin on DNA synthesis and human microvascular endothelial cell growth.
3H-Thymidine incorporation is used to determine the effect of the phosphoric acid agents and suramin on DNA synthesis in HMEC-1 and HMVEC-d cells. Logarithmically growing HMEC-1 or HMVEC-d cells are seeded at 2 X 104 cells/well in six well plates (Falcon) containing 2 ml of MCDB-131 medium supplemented with 5% fetal bovine serum (FBS) (Hyclone). Various amounts of suramin (0 - 500 pM) and equimolar concentrations of the phosphoric acid agents are added to different wells, and the plates incubated for 24 hr.
3H-Thymidine (ICN Radiochemicals) is added and incorporation allowed to proceed for an additional 30 min. After removal of medium, the cell layer is washed twice with 1 ml of cold Hanks balanced salt solution and the cells are dislodged by trypsinization.
The cells are collected in microcentrifuge tubes and washed twice with 1 ml of cold phosphate-buffered saline, and then 1 ml of cold i0% trichloroacetic acid are added. Acid precipitable radioactivity are collected on a glass fiber filter (Whatman Grade GF/C) and the radioactivity is determined in a liquid scintillation spectrometer (Packard).
The effect of various concentrations of suramin and the phosphoric acid agents on cell growth and ongoing DNA synthesis is measured. The inventors found that concentrations of suramin up to 100 p,g/ml did not have any significant inhibitory effect on HMEC-l and porcine pulmonary artery macrovascular endothelial cell growth. As a matter of fact, a stimulatory effect on cell growth with suramin at 50-100 ~g/ml was detected. However, at concentrations higher than 250 p,g/ml, there was a significant dose-related reduction in total protein and total DNA.
The phosphoric acid agents always showed potent reduction in total protein and total DNA even at lower concentrations, suggesting, once again, a different and specific mode of action on endothelial cells by the agents.
The time course for inhibition of protein synthesis by the phosphoric acid agents in human microvascular endothelial cells.
SUBSTITUTE SHEET (RULE 26) 3H-Leucine incorporation is used to determine the effect of the phosphoric acid agents and suramin on protein synthesis in HMEC-1 and HMVEC-d cells. HMEC-1 or HMVEC-d cells are grown to confluence in flasks (P 100) and IC50 doses of the phosphoric analogues or suramin are added for time periods of 6 to 36 hr. For each time period, six replicate flasks are set up. The volume of the medium MCDB-131 is 10 ml per flask. After incubation, the medium was replaced with fresh MCDB-131 containing 0.37 MB ml of 3H-leucine and incubated for a further 90 min. TritonX-100 (240 pl, 10%) were then added to the contents of each flask and agitated.
Trichloroacetic acid (TCA) (4 ml, 20%) is added, the contents mixed and the flasks left overnight at room temperature. After 24 hr, the flask contents was suspended by shaking, transferred to tubes and centrifuged at 43008. The pellets were resuspended in 10% TCA and recentrifuged and then resuspended in 3.0 ml of 0.1 M NaOH. Half of this volume was transferred to scintillation vials in 10 ml of Emulsifier Safe scintillation fluid (Packard) while the remaining 1.5 ml was used to assay DNA content (LaBarca and Paigen, 1980). The radioactive counts due to protein synthesized in the presence of 3H-leucine was normalized to counts/p.g DNA.
Protein synthesis by cells treated with the phosphoric acid agents are expressed relative to that of control cells as the percent of inhibition for each time of incubation.
Comparison of 3H-leucine incorporation into cellular protein in the presence or absence of suramin and the phosphoric acid agents was used as a measure of the inhibition of protein synthesis. There was no change in the rate of protein synthesis until suramin and the cells have been incubated together for at least 24 hr. It was shown that this is the time necessary for a reasonable amount of suramin to be taken up by HMEC-1 cells (Gagliardi et al., 1996). After this period of time, the rate steadily declines. The inhibition of protein synthesis by the phosphoric acid agents was significant after a short time of incubation because these compounds are smaller and less charged than suramin.
Determination of the sensitive phase of the cell cycle for the inhibitory effects of the phosphoric acid agents and suramin on human microvascular endothelial cells.
Suramin has been shown to inhibit cell cycle progression at different phases in various cancer cell lines. There is no data available on the effect of the phosphoric acid agents and SUBSTITUTE SHEET (RULE 26) suramin on cell cycle in human microvascular endothelial cells. Jindal et al.
(1990) first described the inhibitory action of suramin on DNA synthesis and proposed that it was due to a direct action on cellular DNA polymerases. Data from in vitro studies suggest that the optimal benefit from suramin may require prolonged exposure time. It has been reported that prostate carcinoma cells, treated in vitro with suramin, are slowly arrested in the G1 phase. Cell arrest in the G1 phase became evident only after 24 hr of exposure and suramin also induced a decrease in cells in the S phase (Qiao et al., 1994). Suramin inhibited proliferation of human cerebral meningioma cells and increased the percentage of cells in the S and G2/M phase of the cell cycle.
As suramin simultaneously decreased the proliferation rate shown by direct cell counting and 3H-thymidine uptake, the effect in the G2/M phase cannot be attributed to increased proliferative activity.
Consequently, suramin must lead to a prolongation of the S and G2/M phases or to an arrest during these cell cycle phases. However, the data showed that the human microvascular endothelial cells are much more sensitive to inhibition by both suramin and the phosphoric acid agents than the large vessel endothelium, suggesting a stronger promoting activity of bFGF on microvascular as opposed to macrovascular endothelial cells, as described by Braddock et al.
( 1994).
Effects of the phosphoric acid agents on the transit time of endothelial cells through the cell cycle.
Experiments using flow cytometry are performed to determine the effects of the phosphoric acid agents at different doses and comparing them to the results with suramin.
HMEC-1 and HMVEC-d synchronized cells are used to analyze the effects of selected phosphoric acid agents on the distribution of cells in the various phases of the cell cycle using the propidium iodide method according to Vindelov et al. (1985). Briefly, 104 cells are seeded with MCDB-131. After 24 hr, the medium is replaced with fresh medium containing the test compounds at 0-500 p.M. After exposure times of 12, 24, 48 and 72 hr, the cells are collected by trypsinization, stained with propidium iodide and analyzed by flow cytometry for the percentage of cells in GO/Gl, S, and G2/M phase.
SUBSTITUTE SHEET (RULE 25) WO 99/05148 PCTlUS98/15470 These studies compare the effects of the phosphoric acid agents on the percentage of cells in different phases of the cycle for human microvascular endothelial cells.
These results enable us to understand if the same mechanisms are involved in the inhibition of endothelial cell growth by phosphoric acid agents and suramin.
Determination of the involvement of cell signaling pathway components, protein lcinase C
and p34CDC2 lunase, on the inhibition of human microvascular endothelial cell growth 6y the phosphoric acid agents.
Many studies have unraveled the importance of cell signaling pathways in the inhibitory action of certain compounds. Protein kinase C (PKC) is involved in apoptosis induction by some compounds, and suramin has been shown to inhibit PKC. Suramin inhibits PKC
type I-III
activity in a concentration-dependent manner with an ID50 = 50 pM. The inhibition of cyclic AMP-dependent protein kinase activity was much less sensitive to suramin with an IC50 = 656 p,M (Mahoney et al., 1990). Similar inhibitory effects were observed with M-kinase, the constitutively active catalytic fragment of PKC, and autophosphorylation of PKC types I-III.
PKC consists of a family of gene products in animal tissues composed of at least ten distinct proteins (alpha, beta, gamma, delta, epsilon, eta, theta, zeta, iota and mu) that are important regulatory elements in signal transduction, cellular regulation and tumor promotion. It has been shown that endothelial cell proliferation in response to bFGF is dependent upon activation of PKC (Kent et al., 1995) and that activation of PKC is both necessary and sufficient for attachment, spreading and migration of human endothelial cells (Yamamura et al., 1996). The distribution of PKC isotypes is cell specific. The discovery of these isotypes that have distinct physiologic functions provides an explanation for the plethora of cellular events that are mediated by PKC. Investigation of PKC isotypes in human umbilical vein macrovascular endothelial cell (HLTVEC) with Northern and Western blot analyses demonstrated the presence of PKC alpha, delta, epsilon, eta, theta and zeta (Hailer et ai., 1996). Endothelial cell stimulation by bFGF has been well documented to increase cytosolic and perinuclear PKC alpha and epsilon immunoreactivity. These same PKC isotypes were markedly down regulated after prolonged treatment with phorbol esters (Yamamura et al., 1996).
SUBSTITUTE SHEET (RULE 26) The down regulation of PKC alpha and epsilon inhibited endothelial cell migration and proliferation. These findings suggest that PKC alpha and epsilon are the isotypes involved with cytoskeleton events, endothelial cell migration and the proliferative response to bFGF. Because of the importance of PKC in regulating pleiotropic biological processes, it is of importance to 5 identify the inhibition of PKC by the phosphoric analogues in endothelial cells.
Effects of the phosphoric acid agents on PKC activity, in situ, in human microvascnlar endothelial cells in culture.
10 HMEC-1 or HMVEC-d cultures are grown to confluence on gelatinized 96-well plates.
After a 24-hr incubation with the MCDB-131 culture medium containing 5% FBS, cells are washed with MCDB-131 only and then incubated with the experimental media.
Several experiments are necessary to understand the role of the phosphoric acid agents on PKC.
Confluent cells are treated with bFGF (10 ng/ml) in the presence or absence of suramin (100, 200, 1 S 400 and 800 ~g/ml) or equimolar concentrations of the phosphoric acid agents for 5-10-30-60 and I20 minutes. Confluent cultures are treated with the active phorbol esters, PMA and PDD
(10-200 ng/ml), and the inactive analogue, 4-a-PDD (Montesano and Orci, 1985) in the presence or absence of suramin or selected phosphoric acid analogues. A specific inhibitor for PKC, RO-318220, is also used in a similar manner as the control. After the indicated time points, the 20 cells are washed with cold PBS and PKC is assayed with (Ac-MBP(4-14)), which acts as specific substrate of PKC (Koide et al., 1992). To each well, 100,1 total volume of the following are added: lysis buffer (final concentration: 0.137 mM NaCI, 5.4 pM KCI, 0.3 pM
Na3P04, 0.4 pM
K2HPO4, 1 mg/ml glucose, 20 mM HEPES, 10 mM MgCl2, 50 p.g/ml digitonin and 25 mM
B-glycerophosphate, pH 7.2), 100 pM (gamma32P) ATP, 2.3 mM CaCl2, 2p,g/ml 25 phosphatidylserine, and 100 uM Ac-MBP. After incubation, 50 p,l from each well are spotted onto phosphocellulose disks, washed with 1 % concentrated H3P04 in water, and counted. Results are expressed as the percentage of inhibition in comparison to cultures treated with bFGF or active phorbol esters.
SUBSTITUTE SHEET (RULE 26) The phosphoric acid agents show a strong inhibitory effect on PKC activity and this effect can be counter balanced by the addition of active phorbol esters. A clear inhibition of PKCa, the isotype that seems to play the most important role in endothelial cells.
~PL~ 18 PKC isotypes involved in induction of apoptosis in microvascular endothelial cells.
Several experiments are necessary to understand which isotypes of PKC respond to cell growth and apoptosis. The expression of different PKC isotypes are determined in subconfluent cultures, confluent quiescent cultures, subconfluent cultures treated with bFGF or VEGF, subconfluent cultures treated with each of the phosphoric acid agents and subconfluent cultures treated with bFGF or VEGF. Subconfluent cultures are also treated with the active phorbol esters (10-200 ng/ml), PMA and PDD and the inactive analog, 4a-PDD (control) (Montesano and Orci, 1985). HMEC-1 and HMVEC-d cells are also treated with the phorbol esters in the presence or absence of the various phosphoric acid analogues. A specific inhibitor for PKC, RO-318220, are also used in a similar manner with and without the phosphoric acid analogues (Tsopanoglou, 1994).
The total RNA is extracted and the mRNA for the specific isotypes is determined by Northern blots as described by Mattila et al. (1994). The protein for each specific PKC isotype is separated and determined by Western blot analysis (Mattila et al., 1994). The measure of apoptosis is carried out in dishes treated in the same fashion as described above. Cells are analyzed for apoptosis as described.
The conventional PKC isotypes (alpha, beta, gamma) are calcium and phospholipid dependent whereas the novel isotypes (delta, epsilon, eta and theta) do not require calcium for activation. Zeta is both calcium and phorbol ester independent. The isotypes have not been reported in any human microvascular endothelial cells. However, for the rat macrovascular and human macrovascular cells, only alpha and epsilon appear to be involved in growth. The experiments show which PKC isotypes are stimulated by phorbol esters or are inhibited by the phosphoric acid analogues. A major goal is to determine if these PKC isotypes can overcome their inhibition by phosphoric acid analogues when treated with phorbol esters. This would SUBSTITUTE SHEET (RULE 26) suggest that PKC is a major pathway for the induction of apoptosis in the human microvascular endothelial cells.
Effects of the phosphoric acid agents on p34CDC2 kinase activity in human microvascular endothelial cells in cultnre.
CDC2 kinase is the key enzyme controlling G2-M transition in human cells and its inactivation results in cell cycle interruption and G2 block (Bojanowski et al., 1994). In studies using DNA flow cytometry, suramin inhibited meningioma cell proliferation in five different tumor lines by arresting cells in G2-M and S phases of the cell cycle (Schrell et al., 1995). These effects were found under serum-containing and serum-free culture conditions, and in the absence or presence of estradiol or insulin-like growth factor-1. Prolonged exposure (48 hr) to suramin caused an accumulation of MCF-7 human breast cancer cells in the G2-M phase of the cell cycle (Foekens et al., 1993). Suramin has a direct inhibitory effect on purified cdc2 kinase and also modulates the tyrosine phosphorylation of cdc2 kinase in extracts from human small cell lung cancer cells, suggesting that suramin might have a double inhibitory effect on cdc2 kinase in vivo:
one blocking the kinase activity and the second, protecting the tyrosine phosphorylation of the enzyme. CDC kinase was found to be important in cell proliferation, and suramin was reported to influence this kinase as well.
The effects of selected phosphoric acid agents on the p34cdc2-related kinase activity are carried out essentially as described by Bojanowski et al. ( 1994).
Cytoplasmic and nuclear extractions: 100 million cells (HMVEC-1 or HMEC-d) are washed twice with cold PBS and incubated in hypotonic phosphate buffer for 45 min on ice. The cells are then disrupted with a Dounce homogenizes and nuclei separated from the cytopiasmic fraction by centrifugation and extensive washing with hypotonic buffer. The nuclei are incubated for 30 min in the presence of 350 mM NaCI and the nonsoluble nuclear material removed by ultracentrifugation (20 min at 40000 rpm in TL 100 Beckman ultracentrifuge).
The protein concentration are adjusted to 1.5 mg/ml, 20% of glycerol are added and the extracts stored at -20°C.
SUBSTITUTE SHEET (RULE 2fi) pI3-agarose precipitation: extracts (300 p,g protein) or purified p34cdc2 kinase (50 ng protein) are diluted in 400 pl of precipitation buffer in the presence and absence of suramin (0-20-1201tM) or the phosphoric acid agents in equimolar concentrations. After 10 min, 15 p.l of p13-agarose are added and samples incubated at 4°C for 1-3 hr. The samples are subjected to a brief centrifugation, the supernatant eliminated and the precipitates are washed four times in precipitation buffer with vortexing and transferred to a clean Eppendorf tube after the third wash.
The precipitates are used immediately for kinase assays or Western blot.
p34cdc2 kinase assay: 25 ng of purified p34cdc2 kinase or p-13 agarose precipitates are incubated in 20 p,l of kinase buffer, 32P-ATP and p34cdc2 kinase substrate, with or without suramin and the phosphoric acid agents at 25°C for 20 min. Reactions are stopped by placing the samples on ice and spotting 5 ~1 of the reaction mixture onto P81 phosphocellulose filters (Whatman). Filters are washed three times in 50 mM phosphoric acid, dried and the radioactivity retained on the filters are determined by liquid scintillation (Beckman).
Western blot: the precipitates are electrophoresed in 12.5% 12 X 12 cm ready-made polyacrylamide gels (Daichi Co.) and the proteins transferred to Immobilon PVDF membranes (Millipore). Nonspecific binding sites on the membranes are saturated with 5%
skim milk in PBS. The immobilized antigens are revealed using bitinylated secondary antibodies with biotinyl-tyramide/streptavidin reagent (Blast kit, Dupont, USA) to increase the sensitivity of the signal and Nitrotetrazolium/NADH substrate (POD kit, WAKO, Japan) to visualize it. A
Pharmacia LKB image master DTS chromoscan are used for band quantification (Bojanowski et al., 1994).
Suramin inhibits p34cdc2 kinase activity in a dose-related manner and the phosphoric acid agents are also potent p34cdc2 kinase inhibitors. Suramin has been reported to increase the global tyrosine specific phosphorylation of cellular proteins in vivo and the first suramin-sensitive tyrosine phosphatase has recently been described (Ghosh and Miller, 1993). The Western blot shows different electrophoretic mobility p34cdc2 kinase bands between samples treated or not treated with suramin and the phosphoric acid agents, and the immunoblot with anti-phosphotyrosine antibody exhibits a difference in p34cdc2 kinase tyrosine phosphorylation.
SUBSTITUTE SHEET (RULE 26) *rB

Programmed cell death (apoptosis) induced by phosphonic acid agents in human microvascular endothelial cells.
To measure the induction of apoptosis by the phosphonic acid agents, quiescent and exponentially growing endothelial cells are analyzed. After 24 hr of seeding (low cell density) or after confluence is reached (high cell density), the medium is changed with fresh medium containing various amounts of the phosphonic acid agents. The experiments with confluent cultures are carried out also in the presence or absence of bFGF ( 10 ng/ml).
After various exposure times (6-36 hr), the cells are harvested and analyzed for the induction of apoptosis by four different methods:
a) The cells are fixed with 70% ethanol, spread onto microscope slides, stained with acridine orange and analyzed for nuclei (500 cells counted per data point).
b) The cells are fixed in 4% buffered formalin, air dried onto lysine coated slides, quenched for endogenous peroxidases with 2% hydrogen peroxide, treated with terminal deoxyribonucleotide triphosphatase, followed by anti-dioxigenin-peroxidase, DAB substrate solution and finally, methyl green. Cells (100) are counted to determine the percentage of Apotag-positive cells per data point.
c) To demonstrate DNA fragmentation (DNA laddering), harvested cells are treated with protease k and sodium dodecyl sulfate for 12 hr to degrade cellular protein.
The DNA are extracted with phenol/chloroform/isoamyl alcohol, followed by ethanol precipitation. The air dried DNA pellet is re-suspended in TE buffer and run on a 1 % agarose gel for 2 hr at 120 volts.
The gels are stained with ethidium bromide and photographed.
d) To determine the time course of events more exactly and to discover whether the cells enter apoptosis from the GONG 1 stage or S/M stages, a new flow cytometry method established in our Flow Cytometry Core Facility based on the method of Reid et al. (1996) is used. The harvested cells are stained with Hoechst 33342 and merocyanine 540, analyzed by flow cytometry and divided in five groups: viable GO/G1, viable s/G2/M, early apoptotic GO/G1, early apoptotic S/G2/M, and fragmented DNA (late apoptotic) cells.
SUBSTITUTE SHEET (RULE 26) The phosphoric acid agents induce programmed cell death in human micmvascular endothelial cells that are actively proliferating and that the apoptosis process is triggered by cell detachment.
Uptake and intracellular distribution of 3H-phosphoric acid analogues by human microvascular endothelial cells.
We have already shown that 3H-suramin is taken up by human microvascular endothelial cells in culture and that suramin is probably transported by the caveolae system. The suramin 10 incorporation by HMEC-1 cells increased and reached a plateau around 24 hr of incubation, and more than 50% of the suramin taken up by HMEC-1 cells went to the nucleus. Our group is synthesizing various tritiated phosphoric acid agents that are used for experiments in HMEC-1 and HMVEC-d cells. Briefly, tritiated suramin (13 pCi/100 ml of MCDB-131 without FBS) obtained from Moravek Biochemicals (Brea, CA) was incubated at 37°C in 5% C02/air for 15 different periods of time (2-72 hr). Triplicates were carried out for each period of incubation. At the end of each incubation period, the cells were processed through different washings and finally to differential centrifugation. Different cell fractions were transferred to separate scintillation vials, solubilized in a liquid scintillation cocktail and counted in a 2000 CA
TRICARB Liquid Scintillation Counter.
20 The phosphoric acid analogues are taken up by HMEC-I and HMVEC-d cells much faster and in higher amounts than suramin because the phosphoric acid agents are smaller, less charged molecules than suramin and less bound to proteins. The demonstration that the phosphoric acid agents can reach significant intracellular concentrations and its localization is very important for the understanding of the mechanism of action of these compounds.
Effects of phosphoric acid agents on programmed cell death in the chick chorioallantoic membrane (CAM).
The CAM assay has been reported as a suitable model for the demonstration of "in vivo"
induced apoptosis (Brooks et al., 1994). We have shown that suramin and the phosphoric acid SUBSTITUTE SHEET (RULE 28) wo mosias rcT~rs9snsa7o agents are potent inhibitors of angiogenesis in the 6-day CAM assay. To explore the possibility that suramin and/or the phosphoric acid agents induce apoptosis in vascular cells, 6-day old chick embryos are treated with suramin or the phosphoric acid agents (0-200 pM) and injected in the CAM fluid (in the 6-day CAM, the angiogenic vessels grow rapidly embedded with CAM fluid).
After 24, 48 and 72 hr of treatment with the compounds or saline as control, the CAMS are resected for DNA isolation and analysis for oligonucleosomal fragmentation as previously described by Brooks et al., (1994). To identify those cells within the CAM
undergoing apoptosis in response to the treatment with the phosphoric acid agents, cryostat sections prepared from CAMs treated for 24-48 and 72 hr are examined for apoptosis by the Apo-Tag immunoreactivity kit and for endothelial cell specific staining. Co-localization of these markers in the same cells demonstrate that inhibition of angiogenesis by the phosphoric acid agents in vivo in the 6-day CAM assay involves induction of programmed cell death of microvascular endothelial cells.
ExA»
1 S Inhibition effects of phosphoric acid agents on integrins and human microvascular endothelial cell adhesion.
The adhesion receptor integrin, av/i3, has recently been identified as a marker of angiogenic blood vessels in the chick chorioallantoic membrane and in humans (Brooks et al.
1994). It was also apparent that this integrin played a very important role in angiogenesis.
Topical application of a specific antibody against av(33 prevented the growth of new blood vessels in the chick CAM in response to cytokines and fragments of tumors (Brooks et al., 1994).
In vitro, cell interaction with extracellular matrix has been shown to be related to induction of cell proliferation, motility, gene expression and programmed cell death (Ruoslahti and Reed, 1994;
Meredith et al., 1993). In fact, recent studies have shown that ligation of av(33 on human endothelial cells in vitro promotes a rise in calcium and pH (Leavesley et al., 1993), activation of focal adhesion kinase (Defillipi et al., 1994) and the polymerization of the actin cytoskeleton (Sastry and Horwitz, 1993), which in turn regulates cellular shape and motility of endothelial cells on the extracellular matrix. These signaling events triggered by integrins probably play an important role in the functions and survival of microvascular endothelial cells undergoing angiogenesis.
SUBSTITUTE SHEET (RULE 26) wo mosias rcTms9sns4~o It has been shown that prevention of ligation of the av(33 integrin to the extracellular matrix promotes apoptosis of microvascular endothelial cells which have been induced by angiogenic growth factors to enter the cell cycle. After differentiation has occurred and mature blood vessels formed, integrin av(i3 signaling is no longer required for survival of the microvascular endothelial cells and they become refractory to the deleterious effects of the specific integrin antagonists. In conclusion, antagonists of integrin av~i3 disrupt newly forming blood vessels without affecting the preexisting vasculature (Brooks et al., 1994).
We observed that cultures of human microvascular endothelial cells treated with suramin and more clearly with the phosphonic acid agents showed a large number of floating cells after 24 hr of treatment. Similar findings were reported by Pepper et al. (1994) with bovine microvascular endothelial cells treated with suramin, and by Mitchen et al.(1993) with primary epithelial cell cultures from human prostate treated with suramin. When endothelial cells are cultured under conditions that prevent adhesion and spreading, they stop growing, become detached, acquire a round cell shape and enter programmed cell death (Re et al., 1994).
The regulation of integrin function by the urokinase receptor has also been reported (Wei et al., 1996), suggesting that reagents that affect the urokinase receptor can alter integrin function by disrupting the urokinase plasminogen receptor-integrin association representing potential therapeutic agents for tumor invasion and progression. The specific inhibition of the activity of the urokinase receptor mediated cell surface plasminogen activation system by suramin and the direct effect of suramin on the cell surface associated with the urokinase receptor has already been shown (Behrendt et al., 1993; Ellis and Dano, 1993; Pepper et al., 1994).
We postulate that the phosphonic acid agents might interfere with microvascular endothelial cell adhesion to the extracellular matrix, altering integrin functions, causing endothelial cell detachment and consequential induction of apoptosis.
Cell attachment assay: The effects of the phosphonic acid agents in variable amounts (0-200 ~,M) on the interaction of human microvascular endothelial cells with components of the extracellular matrix, such as laminin (interacts with integrin a2~i 1 ), fibronectin (interacts with integrin a5(31) and vitronectin (interacts with integrin av(33), are studied as described by Sriramarao et al. (1993). Briefly, different concentrations of the proteins (0-10 ~.g/ml) diluted in PBS are immobilized on 96 well plates by incubating overnight at 4°C.
The unbound sites are SUBSTITUTE SHEET (RULE 26) *rB

then blocked with serum free medium (MCDB-131 containing 1% BSA and 10 mM
HEPES) by incubating the wells for 1 hr at 37° C. The human microvascular endothelial cells are harvested after washing with PBS and incubating the cells with a PBS-based free enzyme free cell dissociation solution for 30 min at 37°C. The cell suspension are washed with free serum medium and resuspended at 5 X 104 cells/ml and 100 ~1 are added to each well.
The plates are incubated for 1 hr at 37°C. For the anti-integrin antibody (used as positive control), the cells are preincubated for 30 min before being added to the protein coated wells. Plates are washed twice with PBS containing 1 mM calcium and magnesium to remove unbound cells. The adherent cells are fixed with 3.5% paraformaldehyde containing 0.5% crystal violet.
Endothelial cells are gently washed and adherent cells quantitated by measuring the absorbance at 595 nm on a microtiter plate reader.
Human microvascular endothelial cells treated with the active phosphoric acid agents show a decrease in the percentage of cells attached in relation to the controls. This effect is dose-related. Furthermore, we expect to see more significant changes when the plates are coated with vitronectin because its interaction with integrin av(33 is inhibited by polyanions (Panetti et al., 1995). It takes longer than 12 hr for the detection of any suramin toxicity on endothelial cells even at concentrations higher than 200 pM. Hence, any effect observed in this short time incubation experiment can be interpreted as solely due to inhibition of cell attachment to the extracellular matrix by the phosphoric acid agents.
'' 2~4 Effects of the phosphoric acid agents on the production of protease by human microvascular endothelial cells ins vitro.
A tightly controlled increase in extracellular proteolysis, restricted both in time and space, is a very important component of the angiogenic process. This has led to the notion that compounds capable of inhibiting proteolysis could be effective in inhibiting angiogenesis (Pepper et al., 1994). Indeed, protease inhibitors do inhibit angiogenesis and suramin has been shown to alter the proteolytic properties of bovine microvascular endothelial cells.
Suramin has been shown to significantly inhibit plasminogen activator activity induced by bFGF
in fetal bovine aortic endothelial cells at concentrations higher than 250 p,g/ml (Takano et aL, 1994). These SUBSTITUTE SHEET (RULE 26) methods were established in our laboratory and used to study the effect of tamoxifen (a partial antiestrogen) on proteolytic properties of human microvascular endothelial cells. These same methods are used to determine the effects of phosphoric acid agents.
Human microvascular endothelial cells (HMEC-1 and HMVEC-d) are plated in 96 well culture plates. After 24 hr, the medium are replaced with fresh MCDB-131 containing 5% FCS
and varying amounts of the phosphoric acid agents. The experiment is carned out in the presence or absence of 10 ng/ml bFGF. After 18-24 hr of incubation at 37°C with 5% C02/air, the cells are washed and lysed. Total protein is determined in the lysate and 1 pg of total protein is used to determine plasminogen activator (PA) activity with a chromogenic method (American Diagnostics, CT) and with a microplate reader. Human urokinase (Calbiochem, La Jolla, CA) is used to generate the standard curve of PA activity. Results are expressed as the percentage of control activity. Zymographic analysis of PA is performed according to the method of Granelli et al. (1983).
The phosphoric acid agents express inhibitory activity on the proteoiytic properties in a dose-related manner.
Effect of the phosphoric acid agents on cell matrix matalloproteinases (MMPs) in the endothelial cells.
Matrix metalloproteinases are an important group of zinc enzymes responsible for the degradation of the extracellular matrix components, such as collagen and proteoglycans.
Currently, 16 family members have been identified. MMP family member differ from each other structurally by the presence or absence of domains that contribute to activities such as substrate specificity, inhibitor binding, matrix binding and cell surface localization (Powell and Matrisian, 1996). To determine if the phosphoric acid agents were effective inhibitors of the MMP's of endothelial cells, cells were incubated in the presence of the different agents for 48 hours. Tissue samples were taken and subjected to zymography. Tissue samples were placed in glass homogenizers in a three fold weight/volume of lysis buffer consisting of 50 mM
Tris-HCL, pH=7.4, with 200 mM sodium chloride and 0.1% Triton X-100. The samples were homogenized SUBSTTrUTE SHEET (RULE 26) and centrifuged at 1500 g for 20 min. The supernatant was immediately analyzed by zymography. Protein content of each sample was determined by a modified Lowry method.
MMP enzyme activity was detected using polyacrylamide geI eltmphoresis zymography.
SDS-polyacrylamide gel electrophoresis (PAGE) was performed using 8%
acrylamide gels 5 containing 0.1 % gelatin. The volume of test samples loaded was 15 pI.
Electrophoresis was run at 4°C at a constant voltage ( 100 volts). After electrophoresis, gels were incubated in Triton X-100 (2.5%) for 30 minutes to eliminate SDS, prior to being incubated overnight in 50 mM Tris HCL, pH7.5, containing 10 mM CaCl2 at 37°C. The gels were stained in 0.25% (W/v) Coomassie Brilliant Blue and destained in methanol:acetic acid:water (50:40:10). The clear 10 zones in these gels indicates the presence of proteins with gelatinolytic activity. This method allows for identification of pro-metalloproteinases. Migration position of proteins and with standard molecular weight and supernatant from HT 1080 cells that express MMP-2 and MMP-9 were used as controls.
The results with human microvascular endothelial cells are as follows: NF 050, 15 and NF 681 are potent inhibitors of MMP-2 activity. bFGF increased MMP-2 activity in human micmvacular endothelial cells and these phosphoric acid agents inhibited the increase in MMP-2 activity induced by bFGF. These results suggest that the MMP-2 in endothelial cells may be an irnportant component in the angiogenesis process. This inhibition of MMP-2 by the phosphoric acid agents may be an important mechanism for the inhibition of angiogenesis.
Effect of the phosphoric acid agents on cell matrix metalloproteinases (MMP's) in cancer cells in vitro.
For a tumor cell to metastasize, it must break away from its neighbors and penetrate through the surrounding stoma and the basement membrane to enter the circulation. The extensive degradation of the extracellular matrix component in tumor cells depends on the secretion of a battery of metalloendopeptidases that digest a wide range of proteins of the extracellular matrix (Woessner, 1991 ).
Several recent reports have shown a correlation between MMP expression and tumor invasiveness in prostate cancer. Pajough et al. (1991) found that MMP-7 was increased in SUBSTITUTE SHEET (RULE 26) malignant compared to benign prostatic tissue but absent in the stroma. Boag and Young ( 1993) found increased levels of gelatinise A (MMP-2) in malignant prostate and metastatic tissue.
Steams and Wang (1993) analyzed prostrate cancer tissue extracts for gelatinise A (MMP-2) using Northern blot studies. Their results suggested that the enzyme is selectively overexpressed by malignant preinvasive epithelial cells with very low levels in benign tissue and the stroma surrounding the tumor. Wilson et al. (1993) reported gelatinolytic proteinase activities in human prostate secretions, with an increased level of expression in neoplastic compared to benign disease. The results presented by Hamdy et al. (1994) suggest that MMP-9 activity is increased in malignant compared to benign prostatic tissue. The gelatinise B (MMP-9) was not expressed in benign tissue but was detected in 42% of prostate carcinomas. Furthermore, those who exhibited MMP-9 activity in vitro had particularly unfavorable clinical parameters, including well-established prognostic factors, such as high Gleason scores, serum PSA
levels and primary tumor ploidy. Furthermore, 100% of the patients that did not respond to treatment expressed MMP-9.
Metalloproteinases have long been associated with metastasis and are major functional contributors to the metastatic process. MMP's are important contributors to the initial growth of metastasis, regulating access to growth factors from the extracellular matrix and increasing angiogenesis (Chambers and Matrisian, 1997). Activated MMP's are susceptible to inhibition by the general serum proteinase inhibitor, a-2-macroglobulin, and by a family of specific tissue inhibitors of metalloproteinases (TIMP). The two major members of this family, TIMP-1 and TnVIP-2, are expressed by a variety of cell types. They form non-covalent, stoichiometric complexes with both latent and active MMP. TIMP-1 is associated with progelatinase B and TiMP-2 is associated with progelatinase A. Malignant tumors many times exhibit complex patterns of expression of MMP's and TIMP's and therapeutic intervention might induce changes in this ba.tance.
Using the in vitro protocol described in EXAMPLE 25 above, we have shown that the agents are potent inhibitors of MMP-9 activity in prostate cancer cells (PC-3) cells. MMP-2 activity is inhibited in DU-145 prostate cancer cell lines in vitro by the agents. This finding that the agents are potent inhibitors of MMP's activity clearly indicates an important new therapeutic function for the agents in cancer treatment.
SUBSTITUTE SHEET (RULE 28) To determine if the phosphonic acid agents were effective inhibitors of the MMP's of prostate cancer cells (PC3 and DU-145), cells were incubated in the presence of the different agents for 48 hours. Tissue samples were taken and subjected to zymography.
Tissue samples were placed in glass homogenizers in a three fold weight/volume of lysis buffer consisting of 50 mM Tris-HCL, pH=7.4, with 200 mM sodium chloride and 0.1% Triton X-100. The samples were homogenized and centrifuged at 1500 g for 20 minutes. The supernatant was immediately analyzed by zymography. Protein content of each sample was determined by a modified Lowry method.
The expression of metalloproteinases 2 and 9 and tissue inhibitors of metalloproteinases (TIMP) 1 and 2 in human prostate tumor xenografts in nude mice is determined by the indirect immunoperoxidase method. Briefly, tissue sections are deparaffinized by 100%
xylene and then hydrated with a graded series of ethanol. Frozen sections can also be utilized. The endogenous peroxidase is eliminated by incubation in 3% hydrogen peroxide for 30 minutes, and nonspecific binding of IgG to tissue protein blocked by incubation with 100% normal rabbit serum for 1 hour.
The sections are then reacted with monoclonal anti-human gelatinise A (MMP-2) or B (MMP-9) antibody raised in mouse (Oncogene Research Products, 4°C overnight.
Biotinylated anti-mouse antibody is used as the secondary antibody followed by peroxidase-strepavidin complex. The slides are rinsed three times with PBS after each tetrahydrochloride and hematoxylin used for nuclear staining. Negative controls omitting either the primary or secondary antibodies are used for nonspecific staining. The ratio (%) of immunoreactive cells to total carcinoma cells is measured by counting cells in five different fields at X200. TIMPS's 1 and 2 are detected in frozen sections of the tumors using a monoclonal mouse antibody for human TIMP's and the Mouse Unitect Immunohistochemistry detection kit (Oncogene Research Products, Cambridge, MA).
Effect of the phosphonic acid agents on endothelial cell migration in vitro.
Suramin inhibits multiple control points of angiogenesis, such as angiogenic growth factors binding to endothelial cell surface, endothelial cell migration, proliferation and production of proteases (Coffey et al., 1987, Braddock et al., 1994, Pepper et al., 1994). Migration of SUBSTITUTE SHEET (RULE 26) microvascular endothelial cells is a key step in the angiogenesis process and appears to be more sensitive to suramin inhibition than does endothelial cell proliferation (Takano et al., 1994).
Suramin significantly inhibited endothelial cell migration determined by both the number of cells that migrated and the distance traveled by the cells from the wound edge. The data confirmed that suramin inhibits microvascuiar endothelial cell migration in concentrations above 150 ~g/ml and that the phosphoric acid agents are much more potent inhibitors of endothelial cell migration than suramin (10 to 30 times).
Effect of the phosphoric acid agents on tube formation in endothelial cells in vitro.
Endothelial cell differentiation on Matrigel is a useful in vitro model for the study of certain steps in angiogenesis (Schnaper et al., 1993). The possibility that the phosphoric acid agents inhibit angiogenesis in vitro has been studied using Matrigel, a reconstituted matrix prepared from the Englebreth-Holm- Swarm (EHS) tumor extracellular matrix (Kleinman et al., 1982). When human umbilical vein endothelial cells or bovine microvascular endothelial cells were seeded onto Matrigel, they formed a network of capillary-like structures, mimicking the steps that occur during the formation of new micmvessels. Thus, the culture of endothelial cells on Matrigel serves as a useful model for the study of endothelial cell activity during in vitro angiogenesis.
Although there is a dramatic morphologic change in endothelial cell population involving cell elongation, anastomosis and branching, gene transcription and translation are not required for the regulation of this process (Zimrin et al., 1995). Rather, post-translation-al events are involved since the Matrigel-dependent process could be inhibited by addition of a protein kinase inhibitor.
To measure the effect of the phosphoric acid agents on the ability of tube formation by human microvascular endothelial cells, 200 p,l of Matrigel is plated on a 24 well culture plates.
After the Matrigel is allowed to gel for 30 min at 37°C, 40,000 cells (previously grown on plastic dishes) in 1 ml MCDB-131 medium is plated onto the Matrigel. After 18 and 24 hr, the cultures are fixed, stained and the area of tubes formed on the surface of the Matrigel are quantitated using an image analysis system (Loats Associates, Inc., Westminster, MD). Additional experiments need to be carried out in the presence of various amounts of PMA (40-200 ng/ml) to determine if SUBSTITUTE SHEET (RULE 26) the inhibitory effect of the phosphoric acid agents can be overcome by the stimulation of PKC
with a phorbol ester.
The phosphoric acid agents inhibit tube formation in a dose-dependent manner and that stimulation of PKC by the phorbol ester might overcome the inhibition.
Antiangiogenic effects of the phosphoric acid agents expressed in an in vivo mammalian (mouse) model of angiogenesis.
The phosphoric acid agents inhibit angiogenesis in a simple and rapid in vivo model that allows the ready quantitative assessment of angiogenic and antiangiogenic factors. The method developed by Passaniti et al.(1992) consists of subcutaneously injecting mice with bFGF
embedded in Matrigel in the presence of heparin. Subcutaneous injection of Matrigel plus bFGF
and heparin at the ventral midline achieved optimal and reproducible responses. Sprouts from vessels in the adjacent tissue penetrated the gel within 2 days, connecting it with the external vasculature and reaching a plateau after 4 days, and persisted up to 8 days.
Matrigel forms a solid gel when injected into mice and support a rapid and intense angiogenic reaction in the presence of heparin and bFGF. Matrigel has been used to promote differentiation of endothelial cells into capillary structures in culture, and when utilized as a vehicle in vivo, may enhance the selectivity of endothelial cells entering the gel since basement membranes are not readily crossed by fibroblasts and other cells. Gels supplemented with bFGF and heparin induced intense vascularization. The site of injection and the age of the mice can affect the magnitude of the observed angiogenic response. Angiogenesis is quantitated by image analysis of vessels and by measuring the hemoglobin present in the vessels within the gel.
This approach is used to determine whether the selected phosphoric acid agents can inhibit angiogenesis in vivo using C57B1/6 mice (6 months old). All mice are treated on day 1 of the experimental protocol by injection of 0.2 mi of Matrigel with a dose of heparin plus bFGF
shown to induce intense angiogenesis. The animals are also treated i.p. daily with 2.0, 0.2 and 0.05 nmoles/20 gram body weight of phosphoric acid agents shown to be active antiangiogenic compound in the CAM and HMECI all in vitro. The control group receives daily i.p. injections of physiological saline.
SUBSTITUTE SHEET (RULE 2B) After five days of treatment, the mice are euthanized and dissected.
Photographs are taken of the area around the Matrigel implants and the gel is removed along with a section of the peritoneal lining for support, typically the overlying skin. The Drabkin method (Drabkin reagent kit 525, Sigma, St Louis, MO) is used to measure hemoglobin levels in the implants. Protein 5 content of the supernatant fluid is determined using the BioRad protein assay method. All specimens are fixed in 10% buffered formalin for at least 24 hr, dehydrated, embedded in paraffin and sectioned at 5 micron thickness, deparaffinized, and stained with hematoxylin and eosin.
Selected sections are stained for Factor VIII-related antigen using an immunoperoxidase method.
To measure the total area of neovessels, a computerized digitalyzer, the Optomax image analysis 10 system (Optomax), is used.
The model described by Passaniti et al. (1992) is used as it gives ready quantitative assessment of argiogenesis and is reliable and shown to be useful in testing biological factors and drugs that regulate angiogenesis. Suramin is an effective inhibitor of angiogenesis in vivo as described by Pesenti et al. (1992). Furthermore, the active phosphoric acid agents are 10 to 30 15 times more active than suramin in inhibiting angiogenesis. Moreover, a direct correlation is found between the potency of inhibition of angiogenesis in the CAM assay and in the mouse model for the phosphoric acid analogues.
20 The compounds of the present invention are useful in pharmaceutical compositions for systemic administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, suppositories, sterile parenteral solutions or suspensions, sterile non-parenteral solutions or suspensions oral solutions or suspensions, oiI in water or water in oil emulsions and the like, containing suitable quantities of an active ingredient. Topical application 25 can be in the form of ointments, creams, lotions, jellies, sprays, douches, and the like. For oral administration either solid or fluid unit dosage forms can be prepared with the compounds of Formula I. The compounds are useful in pharmaceutical compositions (wt%) of the active ingredient with a carrier or vehicle in the composition in about 1 to 20% and preferably about 5 to 15%.
SUBSTITUTE SHEET (RULE 26) Either fluid or solid unit dosage forms can be readily prepared for oral administration. For example, the compounds can be mixed with conventional ingredients such as dicalciumphosphate, magnesium aluminum silicate, magnesium stearate, calcium sulfate, starch, talc, lactose, acacia, methyl cellulose and functionally similar materials as pharmaceutical excipients or carriers. A sustained release formulation may optionally be used. Capsules may be formulated by mixing the compound with a pharmaceutical diluent which is inert and inserting this mixture into a hard gelatin capsule having the appropriate size. If soft capsules are desired a slurry of the compound with an acceptable vegetable, light petroleum, or other inert oil can be encapsulated by machine into a gelatin capsule.
Suspensions, syrups and elixirs may be used for oral administration of fluid unit dosage forms. A fluid preparation including oil may be used for oil soluble forms. A
vegetable oil such as corn oil, peanut oil or safflower oil, for example, together with flavoring agents, sweeteners and any preservatives produces an acceptable fluid preparation. A surfactant may be added to water to form a syrup for fluid unit dosages. Hydro-alcoholic pharmaceutical preparations may be used having an acceptable sweetener such as sugar, saccharine or a biological sweetener and a flavoring agent in the form of an elixir.
Pharmaceutical compositions for parenteral and suppository administration can also be obtained using techniques standard in the art.
Another preferred use of the compounds is in a transdennal parenteral pharmaceutical preparation in a mammal such as a human. Accordingly, compositions suitable for administration to these areas are particularly included within the invention. The above parenteral solutions or suspensions may be administered transdermally and, if desired a more concentrated slow release form may be administered. Accordingly, incorporation of the active compounds in a slow release matrix may be implemented for administering transdermally. The compounds may be administered transdermally at about 1 to 20% of the composition and preferably about 5 to 15%
wt% of the active ingredient in the vehicle or carrier.
Transdermal therapeutic systems are self contained dosage forms that, when applied to intact skin, deliver drugs) at a controlled rate to the systemic circulation.
Advantages of using the transdermal routing include: enhanced therapeutic efficacy, reduction in the frequency of dosing, reduction of side effects due to optimization of the blood-concentration versus time SUBSTITUTE SHEET (RULE 26) profile, increased patient compliance due to elimination of multiple dosing schedules, bypassing the hepatic "first-pass" metabolism, avoiding gastrointestinal incompatibilities and providing a predictable and extended duration of activity. However, the main function of the skin is to act as a barrier to entering compounds. As a consequence, transdermal therapy has so far been restricted to a limited number of drugs that possess the desirable physiochemical properties for diffusion across the skin barrier. One effective method of overcoming the barrier function of the skin is to include a penetration enhancer in the formulation of a transdermal therapeutic system.
See Barry, Brian W.: Dermatological Formulations: Percutaneous Absorption (Dekker, New York, 1983); Bronough et al, Percutaneous Absorption, Mechanisms-Methodology-Drug Delivery, (Marcel Dekker, New York, NY 1985); and Monkhouse et al, Transdermal drug deliver-problems and promises. Drug Dev. Ind. Pharm., 14, 183-209 (1988).
A penetration enhancer is a chemical compound that, when included in a formulation, temporarily increases the permeability of the skin to a drug allowing more of the drug to be absorbed in a shorter period of time. Several different types of penetration enhancers have been reported such as dimethylsulfoxide, n-decyl methyl sulfoxide, N,N-dimethylacetamide, N,N-dimethyiformamide, 1-dodecylazacycloheptan-2-one (Atone), propylene glycol, ethanol, pyrrolidones such as N-methyl-2-pyrrrolidone (NMP) and surfactants. See Bronough et al, supra, and Stoughton et al, Atone: a New Non-toxic enhancer of percutaneous penetration. Drug Dev.
Inc. Pharm., 9, 725-744 (1983).
N-methyl-2-pyrrolidone is a versatile solvent which is miscible with water, ethyl alcohol, ether, chloroform, benzene, ethyl acetate and carbon disulfide. N-methylpyrrolidone has been widely used as a solvent in industrial processes such as petroleum refining, GAF Corp.: "M-Pyrol (N-methyl-2-pyrrolidone) Handbook.", GAF Corp., New York, 1972. It is currently used as a solubiliting agent in topical and parenteral veterinary pharmaceuticals and is now under 25' consideration for use in products intended for humans, Wells, D.A. et al:
Disposition and Metabolism of Double-Labeled [3H and '4C] N-methyl-2-pyrrolidone in the Rat.
Drug Met.
Disps., 16, 243-249 (1988). Animal and human experiments have shown very little irritation or sensitization potential. Ames type assays and chronic exposure studies have not revealed any significant toxicity, Wells et al, Mutagenicity and Cytotoxicity of N-methyl-2-p[yrrolidone and 4-(methyl amino) Butanoic Acid in the Salinonella/microsome Assay. J. Appl.
Tox., 8, 135-139 SU6STITUTE SHEET (RULE 26) (1988). N-methylpyrrolidone has also been shown to be an effective penetration enhancer. Barry et al, Optimization and Bioavailability of Topical Steroids: Penetration Enhancers Under Occlusion. J. Inv. Derm., 82, 49-52 (1984); Akter et al, Absorption Through human Skin of Ibuprofen and Flurbiprofen; Effect of Dose Variation, Deposited Drug Filins, Occlusion and the Penetration Enhancer N-methyl-2-pyrrolidone. J. Pharm. Pharmacol., 37, 27-37 (1984);
Holegaard et al, Vesical Effect on Topical Drug Delivery IV. Effect of N-methylpyrrolidone and Polar Lipids on Percutaneous Transport. Int. J. Pharm., 43, 233-240 (1988);
Sugibayashi et al, Effect of Several Penetration Enhancers on the Percutaneous Absorption of Indomethacin in Hairless Rat. Chem. Pharm. Bull., 36, 1519-1529 (1988); Bennett et al, Optimization of Bioavailability of Topical Steroids: Non-occluded penetration Enhancers Under Thermodynamic Control. J. Pharm. Pharmacol., 37, 298-304 (1985); Sasaki et al, Enhancing Effect of Pyrrolidone Derivatives on Transdenman Drug Delivery. 1. Ing. J. Pharm., 44, 14-24 (1988);
Lee et al, Toxicity of N-methyl-2-pyrrolidone (NMP): Teratogenic, Subchronic and Two-year Inhalation Studies, Fund. Appl., Tox., 9, 222-235 (1987}.
The above and other drugs can be present in the reservoir alone or in combination form with pharmaceutical carriers. The pharmaceutical carriers acceptable for the purpose of this invention are the art known carriers that do not adversely affect the drug, the host, or the material comprising the drug delivery device. Suitable pharmaceutical carriers include sterile water;
saline, dextrose; dextrose in water or saline; condensation products of castor oil and ethylene oxide combining about 30 to about 35 moles of ethylene oxide per mole of castor oil; liquid acid;
lower alkanols; oils such as corn oil; peanut oil, sesame oil and the like, with emulsifiers such as mono- or di-glyceride of a fatty acid, or a phosphatide, e.g., lecithin, and the like; glycols;
polyalkylene glycols; aqueous media in the presence of a suspending agent, for example, sodium carboxymethylcellulose; sodium alginate; poly(vinylpyrolidone); and the like, alone, or with suitable dispensing agents such as lecithin; polyoxyethylene stearate; and the like. The carrier may also contain adjuvants such as preserving stabilizing, wetting, emulsifying agents and the like together with the penetration enhancer of this invention.
The effective dosage for mammals may vary due to such factors as age, weight activity level or condition of the subject being treated. Typically, an effective dosage of a suramin SUBSTITUTE SHEET (RULE 26) compound is about 12 g administered for 6 weeks (NCI). The phosphoric acid agents may be administered in a dosage of about 3g for 6 weeks.
Compounds of the present invention may be administered topically at about 1 to 20 wt%
of the composition, and preferably about 5 to 15 wt%. Suramin is presently given by sterile i.v.
injection because of the poor absorption from the gut. For suramin treatment of prostate cancer (Stein 1989), suramin is given i.v. (1-2 g/wk) for a 6 week treatment period.
The chemical characteristics of the phosphoric acid agents suggest that higher effective dosages are achievable.
SUBSTITUTE SHEET (RULE 26) Ades EW, Candal FJ, Swerlick RA, et al. Establishment of an immortalized human microvascular endothelial cell line. J. Invest. Dermatol., 99:683, 1992.
Ausprunk DH, DR Knighton, J Folkman. Differentiation of vascular endothelium in the 5 chick chorioallantois: a structural and autoradiographic study. Dev. Biol.
38:237-248, 1974.
Baghdiguian S, JA Boudier, JL Boudier, J Fantini. Autoradiographic localization of tritiated suramin in polarized human colon carcinoma cells. Cancer Lett.
75:151-156, 1993.
Behrendt N, E Ronne, K Dano. Binding of the urokinase-type plasminogen activator to its cell surface receptor is inhibited by low doses of suramin. J Biol Chem 268:5985-5989, 1993.
10 Bojanowski K, S Lelievre, J Markovits, J Couprie, A Jacquemin-Sablon, AK
Larsen.
Suramin is an inhibitor of DNA topoisomerase II in vitro and in Chinese hamster fibrosarcoma cells. Proc. Natl. Acad. Sci. USA 89:3025-3029, 1992.
Bojanowski K, O Filhol, C Cochet, EM Chambaz, AK Larsen. DNA topoisomerase II
and casein kinase II associate in a molecular complex that is catalytically active. J Biol Chem 15 268:22920-22926, 1993.
Bojanowski K, K Nishio, M Fukuda, AK Larsen, N Saijo. Effect of suramin on p34cdc2 kinase in vitro and in extracts from human H69 cells: evidence for a double mechanism of action.
Biochem Biophys Res Commun 203:1574-1580, 1994.
Bos OJM, ELM Vansterkenburg, JP Boon, MJ Fischer, J Wilting, LHM Janssen.
20 Location and characterization of the suramin binding sites of human serum albumin. Biochem Pharmacol., 40:1595, 1990.
Braddock PS, DE Hu, PD Fan, IJ Stratford, AL Hams, R Bicknell. A structure activity analysis of antagonism of the growth factor and angiogenic activity of basic fibroblast growth factor by suramin and related polyanions. Br. J. Cancer 69:890-898, 1994.
25 Braunhut SJ, LS Gudas, J Sasse PA D'Amore. Expression of FGF by F9 teratocarcinoma cells as a function of differentiation. J Cell Biol 108:2467-2476, 1989.
Brooks PC, Montgomery AP, Rosenfeld M, et al. Integrin-a and -a antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell, 79:1157-1164, 1994.
SUBSTITUTE SHEET (RULE 26) Carmichael J, WG DeGraff, AF Gazdar, JD Minna, JB Mitchell. Evaluation of tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing.
Cancer Res 47:936-942, 1987.
Coffey RJ, EB Leof, GD Shipley, HL Moses. Suramin inhibition of growth factor receptor binding and mitogenicity in AKR-2B cells. J. Cell. Physiol. 132:143-148, 1987.
Craig K, S Mi, EO Harrington, JD Chang, S Mallette, JA Ware. Requirement for protein kinase C activation in basic fibroblast growth factor induced human endothelial cell proliferation.
Circ. Res. 77:231-238, 1995.
Crum R, Szabo S, Folkman J. A new class of steroids inhibits angiogenesis in the presence of heparin or a heparin fragment. Science 230:1375-1378, 1985.
Defilippi P, C Bozzo, G Volpe, G Romano, M Venturino, L Silengo, G Tarone.
Integrin-mediated signal transduction in human endothelial cells: analysis of tyrosine phosphorylation events. Cell Adhesion Commun 2:75-86, 1994.
Denekamp J. Vasculature as a target for tumor therapy. In: Hammersen F, Hudlicka O, ed., Progress in Applied Microcirculation. Basel:Karger, 4:28-38, 1984.
Ellis V, K Dano. Specific inhibition of the activity of the urokinase receptor-mediated cell-surface plasminogen activation system by suramin. Biochem J 296:505-510, 1993.
Flamme I, K Schulze-Osthoff, HJ Jacob. Mitogenic activity of chicken chorioallantoic membrane fluid is temporally correlated to vascular growth in the chorioallantoic membrane and related to fibroblast growth factors. Development 11(3):683-690, 1991.
Foekens JA, AM Sieuwerts, EMJ Stuurman-Smeets, HA Peters, JGM Klijn. Effects of suramin on cell-cycle kinetics of MCF-7 human breast cancer cells in vitro. Br J Cancer 67:232-236, 1993.
Folkman J. Angiogenesis - retrospect and outlook. In: Angiogenesis: Key Principles -Science - Technology - Medicine. Ed. by R. Steiner, P.B. Wiesz & R. Larger, Birkhauser Verlag Basel/Switzerland, 1992.
Folkman J. Tumor angiogenesis. Adv. Cancer Res. 43:175-203, 1985.
Gagliardi AR, Hennig B, Collins DC. Antiestrogens Inhibit Endothelial Cell Growth Stimulated By Angiogenic Growth Factors. Anti-Cancer Res., 16:1-6, 1996.
SUBSTITUTE SHEET (RULE 26) Gagliardi AR, H Hadd, DC Collins. Inhibition of angiogenesis by suramin.
Cancer Res.
52:5073-5075, 1992.
Ghosh J, RA Miller. Suramin, an experimental chemotherapeutic drug, irreversibly blocks T cell CD45-protein tyrosine phosphatase in vitro. Biochem Biophys Res Commun 194:36-44, 1993.
Haller T, P Dietl, P Deetjen, H Volkl. The lysosomal compartment as intracellular calcium store in MDCK cells: a possible involvement in InsP3-mediated Ca2=
release. Cell Calcium 19:157-165, 1996.
Jindal HK, CW Anderson, RG Davis, JK Vishwanatha. Suramin affects DNA
synthesis in HeLa cells by inhibition of DNA polymerases. Cancer Res 50:7754-7757, 1990.
Kent KC, S Mii, EO Harrington, JD Chang, S Mallette, JA Ware. Requirement for protein kinase C activation in basic fibroblast growth factor-induced human endothelial cell proliferation. Circ Res 77:231-238, 1995.
Koide H, K Ogita, U Kikkawa, Y Nishizuka. Isolation and characterization of the E
35 subspecies of protein kinase C from rat brain. Proc Natl Acad Sci USA
89:1149-1153, 1992.
LaBarca C, K Paigen. A simple, rapid and sensitive DNA assay procedure. Anal Biochem 102:344-352, 1980.
La Rocca RV, CA Stein, R Danesi, et al. Suramin, a novel antitumor compound. J
Steroid Biochem Molec Biol 37:893-898, 1990.
Leavesley PI, MA Schwartz, M Rosenfeld, DA Cheresh. Integrin al- and a3-mediated endothelial cell migration is triggered through distinct signaling mechanisms.
J Cell Biol 121: i 63-170, 1993.
Mahoney CW, A Azzi, K-P Huang. Effects of suramin, an anti-human immunodeficiency virus reverse transcriptase agent, on protein kinase C. J Biol Chem 265:5424-5428, 1990.
Mattila P, M-L Majuri, S Tiisala, et al. Expression of six protein kinase C
isotypes in endothelial cells. Life Sci 55:1253-1260, 1994.
Meredith JE, B Frazeli Jr, MA Schwartz. Adhesion to fibronectin stimulates isositol lipid synthesis and enhances PDGF-induced inositol lipid breakdown. J Cell Biol 121:673-678, 1993.
Middaugh CR, H Mach, CJ Burke, DB Volkin, JM Dabora, PK Tsai, MW Bruner, JA
Ryan, KE Marfia. Biochemistry 31:9016-9024, 1992.
SU6STITUTE SHEET (RULE 26) WO 99/05148 PCTlUS98/15470 Mitchen J, R Rago, G Wilding. Effects of suramin on the proliferation of primary epithelial cell cultures derived from normal, benign hyperplastic and cancerous human prostates.
Prostate 22:75-89, 1993.
Montesano R, Orci L. Tumor-promoting phorbol ester induced angiogenesis in vitro.
Ce1142:469-472, 1985.
Panetti TS, SA Wilcox, C Horzempa, PJ Mc-Keown-Longo. ava5 Integrin receptor-mediated endocytosis of vitronectin is protein kinase C-dependent. J
Biol Chem 270:18593-18597, 1995.
Passaniti A, RM Taylor, R Pili, Y Guo, PV Long, JA Haney, RR Pauly, DS Grant, GR
Martin. Methods in laboratory investigation. A simple, quantitative method for assessing angiogenesis and antiangiogenic agents using reconstituted basement membrane, heparin, and fibmblast growth factor. Lab Invest 67:519, 1992.
Pepper MS, J-D Vassalli, JW Wilks, L Schweiger, L Orci, R Montesano.
Modulation of bovine microvascular endothelial cell proteolytic properties by inhibitors of angiogenesis. J. Cell Biol. 55:419-434, 1994.
Pesenti E, F Sola, N Mongelli, M Grandi, F Spreafico. Suramin prevents neovascularization and tumor growth through blocking of basic fibroblast growth factor activity.
Br. J. Cancer 66:367-372, 1992.
Pollak M and M Richard. Suramin blockade of insulin like growth factor 1 stimulated proliferation of human osteosarcoma cells. J Natl Cancer Inst. 82:1349-1352, 1990.
Qiao L, JG Pizzolo, MR Melamed. Effects of suramin on expression of proliferation associated nuclear antigens in DU-145 prostate carcinoma cells. Biochem Biophys Res Commun 201:581-588, 1994.
Re F, A Zanetti, M Sironi, N Polentarutti, L Lanfrancone, E Dejana, F Colotta.
Inhibition of anchorage-dependent cell spreading triggers apoptosis in cultured human endothelial cells. J
CeII Biol 127:537-546, 1994.
Reid S, Cross R, Snow EC. Combined Hoechst 33342 and merocyamine 540 staining to examine marine B cell cycle stage, viability and apoptosis. J Immunol Methods 1996, In press.
Ruosiahti E, JC Reed. Anchorage dependence, integrins and apoptosis. Cell 77:477-478, 1994.
SUBSTITUTE SHEET (RULE 26) Sastry SK, AF Horwitz. Integrin cytoplasmic domains: Mediators of cytoskeletal linkages and extra- and intracellular initiated transmembrane signaling. Curr Opin Cell Biol 5:819-831, 1993.
Schnaper HW, Kleinman HK, Grant DS. Roie of laminin in endothelial cell recognition and differentiation. Kidney Int 43:20-25, 1993.
Schrell UM~I, S Gauer, F Kiesewetter, A Bickel, J Hren, EF Adams, R Fahlbusch.
Inhibition of proliferation of human cerebral meningioma cells by suramin:
effects on cell growth, cell cycle phases, extracellular growth factors, and PDGF-BB autocrine growth loop. J
Neurosurg 82:600-607, 1995.
Sriramarao P, M Mendler, MA Bourdon. Endothelial cell attachment and spreading on human tenascin is mediated by a2a1 and ava3 integrins. J Cell Sciences 105:1001-1012, 1993.
Takano S, S Gately, ME Neville, WF Herblin, JL Gross, H Engelhard, M
Perricone, K
Eidsvoog, S Brem. Suramin, an anticancer and angio-suppressive agent inhibits endothelial cell binding of basic fibroblast growth factor, migration, proliferation and induction of urokinase-type plasminogen activator. Cancer Res 64:2654-2660, 1994.
Tsopanoglou NE, GC Haralabopoulos, ME Maragoudakin. Opposing effects on modulation of angiogenesis by protein kinase C and cAMP-mediated pathways. J.
Vasc Res 31:195-204, 1994.
Vindelov LL, IJ Christensen, NI Nissen. A detergent-trypsin method for the preparation of nuclei for flow cytometric DNA analysis. Cytometry 6:348-356, 1985.
Wei Y, M Lukashev, DI Simon, SC Bodary, S Rosenberg, MV Doyle, HA Chapman.
Regulation of integrin function by the urokinase receptor. Science 273:1551, 1996.
Yamamura S, PR Nelson, KC Kent. Role of protein kinase C in attachment, spreading, and migration of human endothelial cells. J Surg Res 63:349-354, 1996.
Zimrin AB, B Villeponteau, T Maciag. Models of in vitro angiogenesis:
endothelial cell differentiation on fibrin but not Matrigel is transcriptionally dependent.
Biochem Biophys Res Cornmun 213:630-638, 1995.
Ausprunk DH, DR Knighton, J Folkman. Differentiation of vascular endothelium in the chick chorioallantois: a structural and autoradiographic study. Dev. Biol.
38:237-248, 1974.
SU6STITUTE SHEET (RULE 28) *rB

SS
The purpose of the above description and examples is to illustrate some embodiments of the present invention without implying any limitation. It will be apparent to those of skill in the art that various modifications and variations may be made to the composition and method of the present invention without departing from the spirit or scope of the invention.
All relevant portions of patents and publications cited herein are incorporated by reference in their entireties ____ _ _ SUBSTITUTE SHEET (RULE 26)

Claims (30)

56
1. A phosphonic acid agent having the formula:
(P-Y n1)m1- Q1-K-(Q2-(Y n2 -P)m2)j wherein:
P is a phosphonic group or a phosphonic salt;
Y is -OCO-, -NR1CO-, or -CON(R1)R2-;
R1 is H, CH2CO2H, or substituted or unsubstituted alkyl;
R2 is substituted or unsubstituted alkyl, aryl, or arylalkyl;
Q1 and Q2 are substituted or unsubstituted aryl groups;
K is H, -NH-CO-NH-, -NH-CS-NH-, -NHCO-R3-CONH-, or -NHCS-R3-CSNH-; provided that when K is H, j is 0;
R3 is a substituted or unsustituted aryl group;
j is 0, 1, or 2;
n1 and n2 are independently 0, 1, or 2; and m1 and m2 are independently an integer from 1 to 4.
2. A phosphonic acid agent according to claim 1 of the following formula:
wherein P is a phosphonic group or a phosphonic salt;
Y is -OCO-, -NR1CO-, or -CON(R1)R2-;
R1 is H, CH2CO2H, or substituted or unsubstituted alkyl;
R2 is substituted or unsubstituted alkyl, aryl. or arylakyl;
R4 and R5 are independently H or a substituted or unsustituted alkyl group;
B is CO, CS, CO-R3-CO, or CS-R3-CS;

R3 is a substituted or unsustituted aryl group;
n1 and n2 are independently 0, 1 or 2; and m1 and m2 are independently an integer from 1 to 4.
3. A phosphonic acid agent according to claim 1 of the following formula:
wherein P is a phosphonic group or a phosphonic group substituted with one or more alkali metals;
Y is -OCO-, -NR1CO-, or -CON(R1)R2-;
R1 is H, CH2CO2H, or substituted or unsubstituted alkyl;
R2 is substituted or unsubstituted alkyl, aryl, or arylakyl;
m1 is an integer from 1 to 4;
n1 is 0, 1, or 2;
R6 is H, substituted or unsubstituted alkyl, or NCOR7; and R7 is aryl, substituted aryl, or nitro substituted aryl.
4. A phosphonic acid agent according to claim 2, wherein B is CO or CS; R4 and R5 are independently H or methyl; Y is -OCO-, -NHCO-, or -CON(CH2CO2H)CH2-, if present; n1 and n2 are independently 0 or 1; m1 and m2 are 1; and P is PO3H2, PO3HM or PO3M2, wherein M is Li, Na, or K.
5. A phosphonic acid agent according to claim 2, wherein B is CO or CS; R4 and R5 are independently H or methyl; Y is -CONH-Ar-, -CONH-Ar-CH2-; n1 and n2 are 1; m1 and m2 are independently 1 or 2; and P is PO3H2, PO3HM or PO3M2, wherein Ar is a substituted or unsubstituted aryl group and M is Li, Na, or K.
6. A phosphoric acid agent according to claim 3, wherein said agent is of the formula:
7. A phosphoric acid agent according to claim 3, wherein said agent is of the formula:
8. A phosphoric acid agent according to claim 1, wherein said agent is of the formula:
9. A phosphoric acid agent according to claim 1, wherein said agent is of the formula
10. A phosphonic acid agent according to claim 1, wherein said agent is of the formula
11. A pharmaceutical composition for the treatment of angiogenesis dependent conditions or tumors comprising an effective amount of a phosphonic acid agent of claim 1 and a pharmaceutically acceptable carrier.
12. A method of treating tumors comprising the step of administering an effective amount of a phosphonic acid agent of claim 1 to a host in need of said treatment.
13. A method of treating angiogenesis dependent conditions comprising the step of administering an effective amount of a phosphonic acid agent of claim 1 to a host in need of said treatment.
14. The method of claim 13, wherein said angiogenesis dependent condition is selected from the group consisting of diabetic retinopathy, arthritis, psoriasis, tumor growth and metastasis.
15. A method for the preparation of a phosphonic acid substituted agent, which method comprises:
reacting an amino-benzenephosphonic acid with a di-acid halide to form the phosphonic acid substituted agent.
16. A method according to claim 15, further comprising:
combining the amino-benzenephosphonic acid with a nitrobenzoyl halide to form a nitrobenzamido-benezephosphonic acid; and reducing the nitrobenzamido-benezephosphonic acid to form an aminobenzamido-benezephosphonic acid prior to the reaction with the di-acid halide.
17. A method according to claim 16, wherein reaction of the amino-benzenephosphonic acid with the nitrobenzoyl halide is conducted in a buffered medium and at a temperature in the range of about 20°C to about 40°C.
18. A method according to claim 16, wherein the reaction of the aminobenzamido-benezephosphonic acid with the di-acid halide comprises dissolving the aminobenzamido-benezephosphonic acid in a buffered aqueous medium, and then combining the buffered solution with the di-acid halide in an organic solvent to form the phosphonic acid substituted agent.
19. A method according to claim 15, wherein the di-acid halide is selected from the group consisting of phosgene, thiophosgene and a dicarboxylic acid halide substituted aryl group.
20. A method according to claim 16, wherein the reducing step comprises hydrogenating the nitrobenzamido-benezephosphonic acid with hydrogen and a catalyst.
21. A method for the preparation of a nitrophenylphosphonic ester, which method comprises:
reacting a nitro-halomethyl benzene, a nitro-phenoxycarbonyl halide, or a nitro-phenylisocyanate with an alkylphosphite to produce the nitrophenylphosphonic ester.
22. A method according to claim 21, wherein the alkylphosphite is selected from the group consisting of P(OCH2CH3)3, P(OCH3)3, HOP(OCH2CH3)2 and HOP(OCH3)2.
23. A method according to claim 21, wherein the halomethylbenzene is combined with trimethylphosphite at a temperature of about 80°C or greater.
24. A method according to claim 21, wherein the nitro-phenoxycarbonyl halide is combined with trimethylphosphite.
25. A method according to claim 21, wherein the nitrophenylisocynate is combined with dimethylphosphite.
26. A method for the preparation of a nitrophenylphosphonic acid, which method comprises:
reacting a nitrophenylphosphonic ester in an anhydrous solvent with a halo-silane at a temperature of about 30°C to about 60°C.
27. A method according to claim 26, further comprising adding sodium iodide to the anhydrous solvent.
28. A phosphonic acid substituted agent that is an inhibitor of metalloproteinase.
29. A pharmaceutical composition for the treatment of angiogenesis dependent conditions or tumors comprising an effective amount of a phosphonic acid agent of claim 28 and a pharmaceutically acceptable carrier.
30. A method of inhibiting metalloproteinase comprising the step of administering an effective amount of a phosphonic acid agent of claim 28 to a host in need of said treatment.
CA002297900A 1997-07-24 1998-07-24 Phosphonic acid naphthylureas and their antiantiogenic and antitumorigenic use Abandoned CA2297900A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/899,966 1997-07-24
US08/899,966 US5831347A (en) 1996-10-09 1997-07-24 Apparatus for determining if the duration of a power failure exceeded predetermined limits
US09/121,124 1998-07-23
US09/121,124 US6096730A (en) 1997-07-24 1998-07-23 Phosphonated agents and their antiangiogenic and antitumorigenic use
PCT/US1998/015470 WO1999005148A1 (en) 1997-07-24 1998-07-24 Phosphonated agents and their antiangiogenic and antitumorigenic use

Publications (1)

Publication Number Publication Date
CA2297900A1 true CA2297900A1 (en) 1999-02-04

Family

ID=31890787

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002297900A Abandoned CA2297900A1 (en) 1997-07-24 1998-07-24 Phosphonic acid naphthylureas and their antiantiogenic and antitumorigenic use

Country Status (1)

Country Link
CA (1) CA2297900A1 (en)

Similar Documents

Publication Publication Date Title
US5736576A (en) Method of treating malignant tumors with thyroxine analogues having no significant hormonal activity
Gibault et al. Non-photoinduced biological properties of verteporfin
US20020132819A1 (en) Novel purinse
AU2001266233B2 (en) Colchinol derivatives as vascular damaging agents
US5922775A (en) Method of treating malignant tumors with ketone thyroxine analogues having no significant hormonal activity
EP1258252B1 (en) Integrin expression inhibitors
US20040019077A1 (en) Protein kinase inhibitors
US7674775B2 (en) Substantially cell membrane impermeable compound and use thereof
US20100160297A1 (en) Compounds for pim kinase inhibition and for treating malignancy
US9962362B2 (en) Use of small molecule inhibitors targeting EYA tyrosine phosphatase
US7115661B1 (en) Methods and compositions for mitigating pain
JP2002533429A (en) Ortho-diphenol compounds, methods for inhibiting PARP and pharmaceutical compositions
US6160166A (en) Phosphonated agents and their antiangiogenic and antitumorigenic use
EP1259520B1 (en) Novel purines
KR20060129504A (en) Decoy nucleic acid to synoviolin gene promoter
CN103974955A (en) Pyrimido- pyridazinone compounds and use thereof
US20210130312A1 (en) Inhibitors of eya3-protein tyrosine phosphatase in dna damage repair signaling of pulmonary arterial hypertension
US6372719B1 (en) ανβ3 integrin antagonists in combination with chemotherapeutic agents
AU739637B2 (en) Phosphonated agents and their antiangiogenic and antitumorigenic use
JP2005501047A (en) Protein kinase inhibitor comprising an ATP mimetic linked to a peptide or peptidomimetic
CN107206012A (en) The method that carcinoma of urinary bladder is treated with Ciclopirox, Ciclopirox Olamine or Ciclopirox prodrug
US6255298B1 (en) Macrophage scavenger receptor antagonists for use in the treatment of cardiovascular diseases
CA2297900A1 (en) Phosphonic acid naphthylureas and their antiantiogenic and antitumorigenic use
Morris et al. Eriochrome Black T, structurally related to suramin, inhibits angiogenesis and tumor growth in vivo
EP1100484A1 (en) Macrophage scavenger receptor antagonists

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead