WO2004062595A2 - Antineoplastic ether lipid compounds with modifications at the sn-1 carbon - Google Patents

Antineoplastic ether lipid compounds with modifications at the sn-1 carbon Download PDF

Info

Publication number
WO2004062595A2
WO2004062595A2 PCT/US2004/000359 US2004000359W WO2004062595A2 WO 2004062595 A2 WO2004062595 A2 WO 2004062595A2 US 2004000359 W US2004000359 W US 2004000359W WO 2004062595 A2 WO2004062595 A2 WO 2004062595A2
Authority
WO
WIPO (PCT)
Prior art keywords
ether lipid
group
ether
compounds
lipid
Prior art date
Application number
PCT/US2004/000359
Other languages
French (fr)
Other versions
WO2004062595A3 (en
Inventor
Walter R. Perkins
Original Assignee
Zeneus Pharma Limited
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
Application filed by Zeneus Pharma Limited filed Critical Zeneus Pharma Limited
Publication of WO2004062595A2 publication Critical patent/WO2004062595A2/en
Publication of WO2004062595A3 publication Critical patent/WO2004062595A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/10Phosphatides, e.g. lecithin

Abstract

Ether lipid compounds of formula (I): pharmaceutically-acceptable salts, prodrugs or isomers thereof are provided, where the variables are as defined herein. The compounds of the invention have antineoplastic activity, and accordingly have utility in treating cancer and related diseases. Enantiomers of these compounds, pharmaceutical compositions, and methods for treating cancer with the pharmaceutical compositions are also provided.

Description

Antineoplastic Ether Lipid Compounds with Modifications at the sn-1 Carbon
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention provides novel ether lipid compounds with
modifications at the sn-1 carbon, pharmaceutically-acceptable salts, prodrugs or
isomers thereof, as well as pharmaceutical compositions, and methods for treating
cancer.
References
The following publications, patents and patent applications are cited in this
application as superscript numbers:
1 Berdel, W.E. , Br. J. Cancer. 64:208-211 (1991).
2 Lohmeyer, M. and Bittman, R., Drugs of the Future. 19:1021-1037
(1994).
3 Berdel, W.E. and Munder, P.G. in Platelet Activating Factor and
Related Lipid Mediators, (F. Snyder, Ed.), pp. 449-468, Plenum Press,
New York, NY (1987).
4 Houlihan, W.J., Lohmeyer, M., Workman, P. and Cheon, S.H. , Med
Res. Rev.. 15:157-223 (1995).
5 Principe, P. and Braquet, P., Rev. Oncol. Hematol.. 18: 155-178 (1995). Berdel, W. E., Bausert, W.R.E., Fink, U. , Rastetter, J. and Munder, P.
G., Anticancer Res. 1. 345-352 (1981).
Bittman, R. in Phospholipids Handbook: Chemical Preparation of
Glycerolipids, (Cevc, G., Ed.), Marcel Dekker, Inc. , New York, NY,
pp. 141-232 (1993), and references therein.
Berdel, W.E., Andreesen, R., and Munder, P.G. in Phospholipids and
Cellular Regulation, Vol 2., (Kuo. Jτ Ed).. CRC Press: Boca Raton, FL,
pp. 41-73 (1985).
Boggs, K.P., Rock, CO. and Jackowski, S., J. Biol. Chem.. 270:
11612-11618 (1995).
Boggs, K.P., Rock, CO. and Jackowski, S., Biochim. Biophys. Acta.
1389:1-12 (1998).
Geilen, C.G., Weider, T., Geilen, C.C., Reutter, W., J. Biol. Chem..
267:6719-6724 (1992).
Gajate, C , Santos-Beneit, A., Modolell, M. and Mollinedo, F., Mol.
Pharmacol, 53:602-612 (1998).
Ruiter, G.A. , Zerp, S.F., Bartelink, H., Van Blitterswijk, W.J. and
Verheij, M., Cancer Res .. 59, 2457-2463 (1999).
Wieder, T., Orfanos, C.E. and Geilen, C.G., J. Biol. Chem., 273:
11025-11031 (1998). Bittman, R. , and Arthur, G. in Liposomes: Rational Design, (A.S.
Janoff. Ed.)f pp 125-144, Marcel Dekker, New York, NY (1998).
Arthur, G. and Bittman, R., Biochim. Biophys. A eta. 1390:85-102
(1998).
Andreseen, R., Osterholz, J., Luckenbach, G.A., Costabel, IL, Schulz,
A., Speth, V. , Munder, P.G. and Lohr, G.W., J. Natl. Cancer lust..
72:53-59 (1984).
Yamamoto, N. and Ngwenya, B.Z., Cancer Res . , 47:2008-2013 (1987).
Heesbeen, E.C, Verdonck, L.F., Hermans, S.W.G., van Heugten,
H.G., Staal, G.E.J., Rijksen, G., FEBS Lett.. 290:231-234 (1991).
Honman, Y., Kasukabe, T., Hozumi, M., Tsushima, S., Nomura, H.,
Cancer Res. , 46:5803-5809 (1980).
Zhou, X., Lu, X., Richard, C, Xiong, W., Litchfield, D.W. , Bittman,
R. and Arthur, G., J. Clin. Invest. 98:937-944 (1996).
Marshall, C.J., CelL 80: 179-185 (1995).
Mayhew, E., Ahmad, I., Bhatia, S., Dause, R., Filep, L, Janoff, A.S.,
Kaisheva, E. , Perkins, W.R. , Zha, Y., Franklin, LC, Biochim.
Biophys. Actar 1329, 139-148 (1997).
Powis, G., Seewald, M.L , Gratas, C, Melder, D., Riebow, L, Modest,
E.L, Cancer Res .. 52:2835- 2840 (1992). Wilcox, R.W., Wykle, R.L., Schmitt, LD. and Daniel, L.W., Lipids.
22:800-807 (1987).
Bishop, F.E. , Dive, C, Freeman, S. and Gescher, A., Cancer
Chemofher. Pharmacol .. 31:85-92 (1992).
Cuvillier, O., Mayhew, E., Janoff, A.S. and Spiegel, S., Blood, 94:
3583-3592 (1999).
Kim, U.T., Bhatia, S.K. and Hajdu, L, Tetrahedron Lett. - 32:6521-6524
(1991).
Ahmad, L, Filep, LL, Franklin, LC, Janoff, A., Masters, G.R.,
Pattassery, J., Peters, Schupaky, LL, Zho, Y. and Mayhew, E., Cancer
Res.. 15:1915-1921 (1997).
Peters, A. P., Ahmad, I., Janoff, A.S. , Pushkerava, M.Y. , Mayhew, E.,
Lipids, 32: 1045-1054 (1997).
Volger, W.R., Whigham, E. , Bennette, W. and Olson, A.C, Exper.
Hemato.. 13:629-633 (1985).
Zheng, B., Ioshi, K., Shoji, M., Eibl, H., Berdel, W.E., Hajdu, L,
Vogler, W.R., Kuo, J.F. , Cancer Res.. 50:3025-3032 (1990).
Kotting, J. and Eibl, H. in Lipases, Their Structure, Biochemistry and
Application, (Peterson, S.B. and Wooley, P. Eds.), pp. 289, Cambridge
University Press, Cambridge, MA (1994). 34 Guivisdalsky, P.N. and Bittman, R., Tetrahedron Lett.. 29:4393-4396
(1988).
35 Abdelmageed, O.S., Duclos, R.I., Abushanab, E., Makriyannis, A.,
Chem. Phys. Lipids. 54:49-59 (1990).
36 Ali, S. and Bittman, R. , Bioche . Cell Biol.. 68:360-365 (1990).
37 Teraji et al. , " Phospholipid Derivatives, and Pharmaceutical
Composition of the Same," U.S. Patent No. 4,562, 179, issued December
31, 1985.
38 Pinchul, A. N., Mistner, B.I. and Shvets, V.I., Chem. Phys. Lipids.
65:65-75 (1993).
All of the above publications, patents and patent applications are herein
incorporated by reference in their entirety to the same extent as if each individual
publication, patent or patent application was specifically and individually indicated
to be incorporated by reference in its entirety.
State of the Art
Alkyllysophospholipids (ALPs) and alkylphosphocholines (APCs) represent
subclasses of potential antitumor agents collectively known as antitumor ether
lipids (AELs). They do not interact with cellular DNA and are therefore not
mutagenic.1"3 The antitumor activities of these compounds, which are based on lysophosphatidylcholine, are now well established; the prototype of the
alkyllysophospholipids (ALPs) , 1 -O-octadecyl-2-O-methyl-glycerophosphocholine
(ET-I8-OCH3), and other ether-linked phosphocholine analogues are in clinical
trials.2' 4"7 Compound ET-18-OCH3, a subclass of alkyl lysophospholipids (ALPs),
is known for its anti-cancer activities against breast (MCF-7), Lewis lung (A549),
ovarian (Ovcar-3) cell lines.2'4 Structurally similar, the platelet activating factor
(PAF) differs from ET-18-OCH3 merely in an ester linkage at the sn-2 position of
the glycerol-backbone. Both PAF and ET-18-OCH3 are known to inhibit protein
kinase C activity and phosphatidylcholine choline biosynthesis.3, 31, 32
ALPs also appear to inhibit the proliferation of tumor cells without
affecting the growth of normal cells.8 While the mechanism of inhibition of cell
proliferation has yet to be resolved, various hypotheses have been proposed. In
some cells, ALPs and APCs appear to induce apoptosis as a consequence of
inhibition of phosphatidylcholine synthesis.9"11 Other theories for the mechanism
of action include activation of the stress activated protein kinase pathways,12"13
drug-induced increase in cellular ceramide levels,14 nutrient starvation, inhibition
of transacylase activity, enhanced lipid peroxidation, inhibition of cellular
signaling pathways15"16 and/or activation of tumoricidal macrophages.17"18
Other studies have revealed that ALPs affect the activity of a large number
of signaling molecules including protein kinase C (PKC), phosphatidylinositol 3-
kinase, phosphatidylinositol-specific phospholipase C, and diacylglycerol kinase.19' 20' 16 Recently another signaling molecule, Raf-1, was added to the list with the
demonstration that ET-18-OCH3 decreased the levels of Raf-1 associating with the
cell membrane in growth-factor stimulated MCF-7 cells which consequently led to
decreased activation of MAP kinase,21 a crucial enzyme required in initiating cell
proliferation.22 It was suggested that Raf-1 is a primary target of ALPs in cells.
The large number of molecules affected by ALPs has complicated the task of
separating their primary site(s) of action from secondary events.
The finding that the glycerol-based ether lipids possess anti-neoplastic
activities, has led investigators to explore isosteric and isoformic analogs of
ET-I8-OCH3 especially in areas of synthesis, biological and biophysical
properties.6"7 Compound ET-18-OCH3, a subclass of alkyl lysophospholipids
(ALPs), is known for its anti-cancer activities against breast (MCF-7), Lewis lung
(A549), ovarian (Ovcar-3) cell lines.2'4 ET-18-OCH3 formulated in liposomes
(ELL- 12), is currently being evaluated in Phase I clinical trial.29"30
Despite the progress that has been made in understanding the underlying
mechanisms of antitumor ether lipids, there remains a need to develop novel
compounds and compositions for the treatment of disease. Ideally, the treatment
methods would advantageously be based on ether lipids that are capable of acting
as anti-neoplastic agents. SUMMARY OF THE INVENTION
The invention is directed to the discovery of a class of anti-tumor ether
lipid compounds having anti-neoplastic activity. Preferably, the invention
provides bioactive either lipid compound with modifications at the sn-1 carbon or
pharmaceutically-acceptable salts, prodrugs or isomers thereof. The invention also
relates to pharmaceutical compositions comprising these compounds, and methods
for treating cancer.
In one embodiment, the invention relates to an ether lipid having formula (I), or a pharmaceutically acceptable salt, isomer or prodrug thereof:
Figure imgf000009_0001
Forrnula (I)
R1 is selected from the group consisting of — CH2CH2(OCH2CH2)rnO~CH3 and -(CH2)nCH=CH(CH2)pCH3.
R2 is selected from the group consisting of —OR3 and
Figure imgf000009_0002
R >3 a „n--d A r R>4 are each independently selected from the group consisting of C alkyl and H; and n, m and p are each independently an integer from 1 to 10.
Preferably, n, m and p are each independently an integer from 1 to 5, more preferably n, m and p are each independently an integer from 1 to 3.
Preferably, R2 is selected from the group consisting of — OMe and
— OSO2Me, or the group consisting of — OEt and — OSO2Et.
Preferred compounds of Formula (I) include:
Figure imgf000010_0001
Figure imgf000010_0002
Figure imgf000010_0003
Preferably, the compound of Formula (I) is optically active, more preferably, the compound of Formula (I) is the D enantiomer. In a preferred embodiment, the compounds according to the invention will
not aggregate platelets (i.e. , mimic PAF). The chemical structure of PAF (platelet
aggregation factor) is shown in Figure 1. In an embodiment of the invention, the
antitumor ether lipid compounds will avoid PAF recognition while maintaining or
enhancing activity and selectivity. However, in those cases where a platelet
aggregation response to the antitumor ether lipid compounds is observed, co-
administration with a PAF antagonist may be used to block such a response. In
yet another embodiment of the invention, the D isomer is used in order to avoid a
platelet aggregation response.
In a further embodiment of the invention, the antineoplastic ether lipid
compounds will (1) inhibit growth of tumor cells, and (2) inhibit growth of
normal cell lines as compared to tumor cells. Further, it is also preferred that the
compounds of the invention will not aggregate platelets, will not lyse red blood
cells and have desirable pharmacokinetic properties.
Additionally, the invention relates to pharmaceutical compositions
comprising a pharmaceutically acceptable carrier and a pharmaceutically effective
amount of a compound of formula (I). The pharmaceutical compositions may
comprise (a) a liposome, emulsion or mixed miscelle carrier and (b) a pharmaceutically effective amount of compound of formula (I) or a
pharmaceutically acceptable salt, isomer or prodrug thereof. The invention further relates to a liposome comprising a compound of formula (I) or a pharmaceutically acceptable salt, isomer or prodrug thereof.
These pharmaceutical compositions can be used in methods for treating a
mammal afflicted with a cancer, comprising administering to the mammal a
therapeutically effective amount of the pharmaceutical composition. Typical
dosages range from about 0.1 to about 1000 mg of the compound of formula (I)
per kg of the body weight of the mammal per day.
The type of cancer to be treated may be selected from the group consisting
of, but not limited to: lung cancers, brain cancers, colon cancers, ovarian cancers, breast cancers, leukemias, lymphomas, sarcomas, and carcinomas.
The treatment methods according to the invention may also include
administering to the mammal an additional biologically active agent. Any suitable
biologically active agent may be used in combination with the ether lipids of the
invention. In a preferred embodiment, the additional biologically active agent may
be selected from the group consisting of antineoplastic agents, antimicrobial
agents, and hematopoietic cell growth stimulating agents.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 depicts the structure of l-O-octadecanol-2-O-methyl-OT-
glycero-3-phosphocholine (ET-18-OCH3) (left), PAF (center) and lyso-PC (right).
PAF differs in structure in that the methoxy (-OCH3) is replaced with an acetyl (-OCOCH3) group; i.e. , the ether linkage at sn-2 is replaced with an ester linkage.
For lyso-PC, the sn-1 linkage is an ester and a hydroxyl group resides at the sn-2
position.
FIG. 2 depicts a general scheme for the synthesis of compounds of the
invention, comprising (a) protecting the sn-3 alcohol, (b) ring opening of the
epoxide with an alcohol, (c) derivatizing the sn-2 alcohol group, (d) deprotecting
the sn-3 alcohol group, (e) reacting the sn-3 alcohol with phosphorus oxy chloride,
and (f) reacting the phosphate with a choline salt/pyridine, followed by water to
give a compound of formula (I).
DETAILED DESCRIPTION OF THE INVENTION
As stated above, this invention relates to novel ether lipid compounds,
pharmaceutically-acceptable salts, prodrugs, or isomers thereof, which have utility
as anti-neoplastic agents. In particular, the invention relates to ether lipid
compounds of formula (I), having modifications at the sn-1 carbon. However,
prior to describing this invention in further detail, the following terms will first be
defined. Definitions
The term "alkyl" refers to saturated aliphatic groups including
straight-chain, branched-chain, cyclic groups, and combinations thereof. The
alkyl groups preferably have between 1 to 20 carbon atoms.
The term "alkenyl" refers to unsaturated aliphatic groups including
straight-chain, branched-chain, cyclic groups, and combinations thereof, having at
least one double bond and having the number of carbon atoms specified. The
alkenyl groups preferably have between 1 to 20 carbon atoms.
The term "cyclic alkyl" or "cycloalkyl" refers to alkyl group forming an
aliphatic ring. Preferred cyclic alkyl groups have about 3 carbon atoms.
The term "direct link" as used herein refers to a bond directly linking the
substituents on each side of the direct link.
The ether lipids of the invention have a 3 carbon alcohol, glycerol, as the
backbone. With the 3 carbons of glycerol, positions are designated as
stereospecific numbers, sn, to distinguish location. The designations ".STΪ-I , " "sn-
2," and "sn-3" identify glycerol carbons 1, 2, and 3, respectively. The glycerol
carbons are labeled below for formula (I):
Figure imgf000015_0001
Formula (I)
"Pharmaceutically acceptable salt" refers to pharmaceutically acceptable
salts that are derived from a variety of organic and inorganic counter ions well
known in the art and include, by way of example only, sodium, potassium,
calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the
molecule contains a basic functionality, salts of organic or inorganic acids, such as
hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the
like. Examples of pharmaceutically acceptable acid addition salts includes salts
which retain the biological effectiveness and properties of the free bases and which
are not biologically or otherwise undesirable, formed with inorganic acids such as
hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid
and the like, and organic acids such as acetic acid, propionic acid, glycolic acid,
pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid,
tartar ic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid,
methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid
and the like. Examples of pharmaceutically acceptable base addition salts include
those salts derived from inorganic bases such as sodium, potassium, lithium,
ammonium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum
bases, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium and magnesium salts. Salts derived from pharmaceutically acceptable
organic nontoxic bases include salts of primary, secondary, and tertiary amines,
substituted amines including naturally occurring substituted amines, cyclic amines
and basic ion exchange resins, such as isopropylamine, trimethylamine,
diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol,
trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine,
hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine,
theobromine, purines, piperizine, piperidine, N-ethylpiperidine, polyamine resins
and the like. Particularly preferred organic nontoxic bases are isopropylamine,
diethylamine, ethanolamine, trimethamine, dicyclohexylamine, choline, and
caffeine.
"Prodrug" means any compound which releases an active parent drug
according to formulas (I) in vivo when such prodrug is administered to a
mammalian subject. Prodrugs of a compound may be prepared by modifying
functional groups present in the compound in such a way that the modifications
may be cleaved in vivo to release the parent compound. Prodrugs include
compounds of formula (I) wherein a hydroxy, amino, or sulfhydryl group is
bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl,
amino, or sulfhydryl group, respectively. Examples of prodrugs include, but are
not limited to esters (e.g. , acetate, formate, and benzoate derivatives), carbamates
(e.g., N,N-dimethylamino-carbonyl), and the like. "Isomers" are compounds that have the same molecular formula but differ
in the nature or sequence of bonding of their atoms or the arrangement of their
atoms in space. Isomers that differ in the arrangement of their atoms in space are
termed "stereoisomers. " Stereoisomers that are not mirror images of one another
are termed "diastereomers" and those that are non-superimposable mirror images
of each other are termed "enantiomers. " An enantiomer can be characterized by
the absolute configuration of its asymmetric center and is described by the R- and
S- sequencing rules of Cahn and Prelog, or by the manner in which the molecule
rotates the plane of polarized light and designated as dextrorotatory or levorotatory
(i.e. , as (+) or (-)-isomers respectively). A chiral compound can exist as either
individual enantiomer or as a mixture thereof. A mixture containing equal
proportions of the enantiomers is called a "racemic mixture".
"Treating" or "treatment" of a disease includes:
(1) preventing the disease, i.e. causing the clinical symptoms of the disease
not to develop in a mammal that may be exposed to or predisposed to the
disease but does not yet experience or display symptoms of the disease,
(2) inhibiting the disease, i.e. , arresting or reducing the development of the
disease or its clinical symptoms, or
(3) relieving the disease, i.e. , causing regression of the disease or its clinical
symptoms. A "therapeutically effective amount" means the amount of a compound
that, when administered to a mammal for treating a disease, is sufficient to effect
such treatment for the disease. The "therapeutically effective amount" will vary
depending on the compound, the disease and its severity and the age, weight, etc. ,
of the mammal to be treated.
A "pharmaceutically acceptable carrier" means an carrier that is useful in
preparing a pharmaceutical composition that is generally safe, non-toxic and
neither biologically nor otherwise undesirable, and includes a pharmaceutically
acceptable excipient that is acceptable for veterinary use or human pharmaceutical
use. A "pharmaceutically acceptable excipient" as used in the specification and
claims includes both one and more than one such excipient. Some examples of
suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches,
gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate,
microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup,
and methyl cellulose. The formulations can additionally include: lubricating
agents such as talc, magnesium stearate, and mineral oil; wetting agents;
emulsifying and suspending agents; preserving agents such as methyl- and
propylhydroxy-benzoates; sweetening agents; and flavoring agents. The
compositions of the invention can be formulated so as to provide quick, sustained
or delayed release of the active ingredient after administration to the patient by
employing procedures known in the art. " Cancer" refers to a group of diseases characterized by uncontrolled
growth and spread of abnormal cells, often resulting in the formation of a
non-structured mass or tumor. Illustrative tumors include carcinomas, sarcomas
and melanomas, such as basal cell carcinoma, squamous cell carcinoma,
melanoma, soft tissue sarcoma, solar keratoses, Kaposi's sarcoma, cutaneous
malignant lymphoma, Bowen's disease, Wilm's tumor, hepatomas, colorectals
cancer, brain tumors, mycosis ftmgoides, Hodgkin's lymphoma, polycythemia
vera, chronic granulocytic leukemia, lymphomas, oat cell sarcoma, and the like.
Tumors may also include benign growths such as condylomata acuminata (genital
warts) and moles and common warts.
An "anti-neoplastic agent" is a pharmaceutical which inhibits or causes the
death of cancer or tumor cells.
An "antimicrobial agent" is a substance that either destroys or inhibits the
growth of a microorganism at concentrations tolerated by the infected host.
A "hematopoietic cell growth stimulating agent" is one that stimulates
blood cell growth and development, i.e. of red blood cells, leukocytes, and
platelets. Such agents are well known in the art. For example, in order to
increase infection-fighting white blood cell production, recombinant
granulocyte-colony stimulating factor may be used to stimulate the growth of neutrophils. Another example of a hematopoietic cell growth stimulating agent is
recombinant granulocyte macrophage-colony stimulating factor, which increases production of neutrophils, as well as other infection-fighting white blood cells,
granulocytes and monocytes, and macrophages. Another hematopoietic agent is
recombinant stem cell factor, which regulates and stimulates the bone marrow,
specifically to produce stem cells.
Compound Preparation
The compounds of formula (I) can also be prepared via several divergent
synthetic routes with the particular route selected relative to the ease of compound
preparation, the commercial availability of starting materials, and the like. For
instance, the compounds of formula (I) may be synthesized and tested using the
methods ememplified in the examples and the instant specification. Such methods
may be further adapted to produce analogs, derivatives and variants within the
scope of formula (I).
Figure 2 shows the steps and reagents required for the synthesis of ether
lipid compounds of formula (I) with substitution at sn-2 carbon product in only a
few steps in good yields. The starting material shown in the scheme may be
optically active (R)-glycidol or (S)-glycidol, depending upon which isomer is
desired.
In step (a), the alcohol group is protected with a suitable protecting group.
Suitable protecting groups are described, for example, in Protective Groups in
Organic Synthesis, Third Edition (Peter G. M. Wuts and Theodora Greene, Editors), Wiley, John & Sons, Incorporated (1999), which is hereby incorporated
by reference in its entirety. In a preferred embodiment, the protecting group is a
benzyl group.
In a preferred embodiment, O-Benzyl glycidyl ether is used as the starting
material. O-Benzyl glycidyl ether has been used to design and synthesize a class
of novel ether lipids with (R)- and (S) -configurations. This procedure can also be
applied to prepare the phospholipids in multi gram quantities, providing an access
to scale up a wide variety of glycero-based lipids in efficient manners. This route
provides a significant improvement over the conventional method, which involves
eight steps from L-serine as a precursor, for the preparation of 2-aza
phosphocholines.28 This is a simple and short procedure to synthesize (R)- and (S)
ether lipids with substitution sn-2 carbon, starting from the commercially available
O-benzylglycidyl ether precursor with high percentage enantiomeric excess.
In step (b), the epoxide is subjected to ring-opening conditions with an
alcohol. As shown in the scheme, this reaction may be carried out by reacting a
suitable nucleophile in CH2C12 with
Figure imgf000021_0001
at room temperature until the
reaction is complete. Preferred nucleophiles include alcohols, particularly, HO-CH2CH2(OCH2CH2)mO-CH3, HO-(CH2)nCH=CH(CH2)pCH3 (cis double bond) and HO-(CH2)nCH=CH(CH2)pCH3 (trans double bond).
In step (c), the alcohol moiety may be converted to a -OR3 or -O-SO2-R4
group. For example, in order to covert the alcohol to a -OR3 group, where R3 is a CM alkyl, the alcohol may be refluxed with 2,6-di-t-butyl-4-methylpyridine with a
suitable alkyling agent in anhydrous methylene chloride at around 60 °C until the
reaction is complete. Examples of suitable alkylating agents include methyl
trifluoromethanesulfonate (methyl triflate) or ethyl trifluoromethanesulfonate (ethyl
triflate).
The alcohol may alternatively be converted to a -O-SO2-R4 group, where
R4 is hydrogen or a C1 alkyl with a suitable sulfonating agent. Examples of
suitable sulfonating agents include methanesulfonyl chloride (mesyl chloride) or
ethanesulfonyl chloride.
In step (d), the protecting group at the sn-3 position may be removed using
suitable deprotecting conditions. For instance, if the protecting group is a benzyl
group, it may be removed by using H2 gas and 5 % Pd on carbon until the reaction
is complete.
In step (e), the alcohol at the sn-3 position is reacted with POCl3 and Et3N.
The reaction is stirred at 0 °C to room temperature for about 3-4 hours or until the
reaction is complete. Next, choline tosylate/pyridine is added and the reaction
proceeds at room temperature until reaction is complete, followed by quenching
with water and stirring at room temperature for about 1 hour.
If an alcohol group is desired at the sn-2 position, the above synthesis may
be modified by protecting the alcohol at the sn-2 position after step (b). This
protecting group selected should be stable under the reaction conditions of steps (d), (e) and (f). The protecting group of the sn-2 alcohol can then be removed at
the end of the synthesis to yield an alcohol.
The desired product is then isolated and purified using any suitable
technique known in the art. For instance, many of the compounds can be easily
purified by column chromatography.
Pharmaceutical Formulations
When employed as pharmaceuticals, the compounds of formula (I) are
usually administered in the form of pharmaceutical compositions. These
compounds can be administered by a variety of routes including oral, rectal,
transdermal, subcutaneous, intravenous, intramuscular, and intranasal. These
compounds are effective as both injectable and oral compositions. Such
compositions are prepared in a manner well known in the pharmaceutical art and
comprise at least one active compound.
This invention also includes pharmaceutical compositions which contain, as
the active ingredient, one or more of the compounds of formula (I) above
associated with pharmaceutically acceptable carriers. In making the compositions
of this invention, the active ingredient is usually mixed with an excipient, diluted
by an excipient or enclosed within such a carrier which can be in the form of a
capsule, sachet, paper or other container. When the excipient serves as a diluent,
it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of
tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions,
solutions, syrups, aerosols (as a solid or in a liquid medium), ointments
containing, for example, up to 10% by weight of the active compound, soft and
hard gelatin capsules, suppositories, sterile injectable solutions, and sterile
packaged powders.
In preparing a formulation, it may be necessary to mill the active
compound to provide the appropriate particle size prior to combining with the
other ingredients. If the active compound is substantially insoluble, it ordinarily is
milled to a particle size of less than 200 mesh. If the active compound is
substantially water soluble, the particle size is normally adjusted by milling to
provide a substantially uniform distribution in the formulation, e.g. about 40
mesh.
Some examples of suitable excipients include lactose, dextrose, sucrose,
sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth,
gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone,
cellulose, sterile water, syrup, and methyl cellulose. The formulations can
additionally include: lubricating agents such as talc, magnesium stearate, and
mineral oil; wetting agents; emulsifying and suspending agents; preserving agents
such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring
agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to
the patient by employing procedures known in the art.
The compositions are preferably formulated in a unit dosage form, each
dosage containing from about 5 to about 100 mg, more usually about 10 to about
30 mg, of the active ingredient. The term "unit dosage forms" refers to physically
discrete units suitable as unitary dosages for human subjects and other mammals,
each unit containing a predetermined quantity of active material calculated to
produce the desired therapeutic effect, in association with a suitable
pharmaceutical excipient. Preferably, the compound of formula (I) above is
employed at no more than about 20 weight percent of the pharmaceutical
composition, more preferably no more than about 15 weight percent, with the
balance being pharmaceutically inert carrier(s).
The active compound is effective over a wide dosage range and is generally
administered in a pharmaceutically effective amount. It will be understood,
however, that the amount of the compound actually administered will be
determined by a physician, in the light of the relevant circumstances, including the
condition to be treated, the chosen route of administration, the actual compound
administered, the age, weight, and response of the individual patient, the severity
of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active
ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present
invention. When referring to these preformulation compositions as homogeneous,
it is meant that the active ingredient is dispersed evenly throughout the
composition so that the composition may be readily subdivided into equally
effective unit dosage forms such as tablets, pills and capsules. This solid
preformulation is then subdivided into unit dosage forms of the type described
above containing from, for example, 0.1 to about 500 mg of the active ingredient
of the present invention.
The tablets or pills of the present invention may be coated or otherwise
compounded to provide a dosage form affording the advantage of prolonged
action. For example, the tablet or pill can comprise an inner dosage and an outer
dosage component, the latter being in the form of an envelope over the former.
The two components can be separated by an enteric layer which serves to resist
disintegration in the stomach and permit the inner component to pass intact into the
duodenum or to be delayed in release. A variety of materials can be used for such
enteric layers or coatings, such materials including a number of polymeric acids
and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and
cellulose acetate.
The liquid forms in which the novel compositions of the present invention
may be incorporated for administration orally or by injection include aqueous
solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as corn oil, cottonseed oil, sesame oil, coconut oil,
or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and
suspensions in pharmaceutically acceptable, aqueous or organic solvents, or
mixtures thereof, and powders. The liquid or solid compositions may contain
suitable pharmaceutically acceptable excipients as described supra. Preferably the
compositions are administered by the oral or nasal respiratory route for local or
systemic effect. Compositions in preferably pharmaceutically acceptable solvents
may be nebulized by use of inert gases. Nebulized solutions may be inhaled
directly from the nebulizing device or the nebulizing device may be attached to a
face mask tent, or intermittent positive pressure breathing machine. Solution,
suspension, or powder compositions may be administered, preferably orally or
nasally, from devices which deliver the formulation in an appropriate manner.
The following formulation examples illustrate representative pharmaceutical
compositions of the present invention.
For ulation Example 1
Hard gelatin capsules containing the following ingredients are prepared:
Quantity
Ingredient (mg/capsule)
Active Ingredient 30.0
Starch 305.0
Magnesium stearate 5.0
The above ingredients are mixed and filled into hard gelatin capsules in 340
mg quantities. Formulation Example 2
A tablet formula is prepared using the ingredients below:
Quantity
Ingredient (mg/tablet)
Active Ingredient 25.0
Cellulose, microcrystalline 200.0
Colloidal silicon dioxide 10.0
Stearic acid 5.0
The components are blended and compressed to form tablets, each
weighing 240 mg. Formulation Example 3
A dry powder inhaler formulation is prepared containing the following
components:
Ingredient Weight %
Active Ingredient 5
Lactose 95
The active ingredient is mixed with the lactose and the mixture is added to
a dry powder inhaling appliance.
Formulation Example 4
Tablets, each containing 30 mg of active ingredient, are prepared as
follows: Quantity
Ingredient mg/tablef)
Active Ingredient 30.0 mg
Starch 45.0 mg
Microcrystalline cellulose 35.0 mg
Polyvinylpyrrolidone
(as 10% solution in sterile water) 4.0 mg
Sodium carboxymefhyl starch 4.5 mg
Magnesium stearate 0.5 mg
Talc - 1.0 mg
Total 120 mg The active ingredient, starch and cellulose are passed through a No. 20
mesh U.S. sieve and mixed thoroughly. The solution of poly vinylpyrrolidone is
mixed with the resultant powders, which are then passed through a 16 mesh U.S.
sieve. The granules so produced are dried at 50° to 60° C and passed through a 16
mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and
talc, previously passed through a No. 30 mesh U.S. sieve, are then added to the
granules which, after mixing, are compressed on a tablet machine to yield tablets
each weighing 120 mg.
Formulation Example 5
Capsules, each containing 40 mg of medicament are made as follows:
Quantity
Ingredient (mg/capsule)
Active Ingredient 40.0 mg
Starch 109.0 mg
Magnesium stearate 1.0 mg
Total 150.0 mg
The active ingredient, starch, and magnesium stearate are blended, passed
through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg
quantities. Formulation Example 6
Suppositories, each containing 25 mg of active ingredient are made as
follows:
Ingredient Amount
Active Ingredient 25 mg
Saturated fatty acid glycerides 2,000 mg
The active ingredient is passed through a No. 60 mesh U.S. sieve and
suspended in the saturated fatty acid glycerides previously melted using the
minimum heat necessary. The mixture is then poured into a suppository mold of
nominal 2.0 g capacity and allowed to cool.
Formulation Example 7
Suspensions, each containing 50 mg of medicament per 5.0 mL dose are
made as follows:
Ingredient Amount
Active Ingredient 50.0 mg
Xanfhan gum 4.0 mg
Sodium carboxymethyl cellulose (11 %)
Microcrystalline cellulose (89%) 50.0 mg
Sucrose 1.75 g
Sodium benzoate 10.0 mg
Flavor and Color q.v.
Purified water to 5.0 mL
The active ingredient, sucrose and xanthan gum are blended, passed through
a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of the
microcrystalline cellulose and sodium carboxymethyl cellulose in water. The
sodium benzoate, flavor, and color are diluted with some of the water and added
with stirring. Sufficient water is then added to produce the required volume. Formulation Example 8
Quantity
Ingredient (mg/capsule)
Active Ingredient 15.0 mg
Starch 407.0 mg
Magnesium stearate 3.0 mg
Total 425.0 mg
The active ingredient, starch, and magnesium stearate are blended, passed
through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 425.0
mg quantities.
Formulation Example 9
A subcutaneous formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 5.0 mg
Corn Oil 1.0 mL
Formulation Example 10
A topical formulation may be prepared as follows:
Ingredient Quantity
Active Ingredient 1-10 g
Emulsifying Wax 30 g
Liquid Paraffin 20 g
White Soft Paraffin to 100 g
The white soft paraffin is heated until molten. The liquid paraffin and
emulsifying wax are incorporated and stirred until dissolved. The active
ingredient is added and stirring is continued until dispersed. The mixture is then
cooled until solid.
Another preferred formulation employed in the methods of the present
invention employs transdermal delivery devices ("patches"). Such transdermal
patches may be used to provide continuous or discontinuous infusion of the
compounds of the present invention in controlled amounts. The construction and
use of transdermal patches for the delivery of pharmaceutical agents is well known
in the art. See, e.g., U.S. Patent 5,023,252, issued June 11, 1991, herein
incorporated by reference in its entirety. Such patches may be constructed for
continuous, pulsatile, or on demand delivery of pharmaceutical agents.
Frequently, it will be desirable or necessary to introduce the pharmaceutical
composition to the brain, either directly or indirectly. Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to
bypass the blood-brain barrier. One such implantable delivery system used for the
transport of biological factors to specific anatomical regions of the body is
described in U.S. Patent 5,011,472 which is herein incorporated by reference in its
entirety.
Indirect techniques, which are generally preferred, usually involve
formulating the compositions to provide for drug latentiation by the conversion of
hydrophilic drugs into lipid-soluble drugs. Latentiation is generally achieved
through blocking of the hydroxy, carbonyl, sulfate, and primary amine groups
present on the drug to render the drug more lipid soluble and amenable to
transportation across the blood-brain barrier. Alternatively, the delivery of
hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic
solutions which can transiently open the blood-brain barrier.
Other suitable formulations for use in the present invention can be found in
Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia,
PA, 17th ed. (1985), which is hereby incorporated by reference in its entirety.
Utility
The compounds and pharmaceutical compositions of the invention are useful
as anti-neoplastic agents, and accordingly, have utility in treating cancer in
mammals including humans. As noted above, the compounds described herein are suitable for use in a
variety of drug delivery systems described above. Additionally, in order to
enhance the in vivo serum half-life of the administered compound, the compounds
may be encapsulated, introduced into the lumen of liposomes, prepared as a
colloid, or other conventional techniques may be employed which provide an
extended serum half-life of the compounds.
The amount of compound administered to the patient will vary depending
upon what is being administered, the purpose of the administration, such as
prophylaxis or therapy, the state of the patient, the manner of administration, and
the like. In therapeutic applications, compositions are administered to a patient
already suffering from cancer in an amount sufficient to at least partially arrest
further onset of the symptoms of the disease and its complications. An amount
adequate to accomplish this is defined as "therapeutically effective dose."
Amounts effective for this use will depend on the judgment of the attending
clinician depending upon factors such as the degree or severity of cancer in the
patient, the age, weight and general condition of the patient, and the like.
Preferably, for use as therapeutics, the compounds described herein are
administered at dosages ranging from about 0.1 to about 500 mg/kg/day.
In prophylactic applications, compositions are administered to a patient at
risk of developing cancer (determined for example by genetic screening or familial
trait) in an amount sufficient to inhibit the onset of symptoms of the disease. An amount adequate to accomplish this is defined as "prophylactically effective dose. "
Amounts effective for this use will depend on the judgment of the attending
clinician depending upon factors such as the age, weight and general condition of
the patient, and the like. Preferably, for use as prophylactics, the compounds
described herein are administered at dosages ranging from about 0.1 to about 500
mg/kg/day.
The compounds of the invention may also be used in combination therapy
with one or more additional biologically active agents. Virtually any suitable
biologically active agent may be administered together with the ether lipids of the
present invention. Such agents include but are not limited to antibacterial agents,
antiviral agents, anti-fungal agents, anti-parasitic agents, tumoricidal agents,
anti-metabolites, polypeptides, peptides, proteins, toxins, enzymes, hormones,
neurotransmitters, glycoproteins, lipoproteins, immunoglobulins,
immunomodulators, vasodilators, dyes, radiolabels, radio-opaque compounds,
fluorescent compounds, receptor binding molecules, anti-inflammatories,
antiglaucomic agents, mydriatic compounds, local anesthetics, narcotics, vitamins,
nucleic acids, polynucleotides, etc. The entrapment of two or more compounds
simultaneously may be especially desirable where such compounds produce
complementary or synergistic effects. In particular, such biologically active agents
include, but are not limited to, antineoplastic agents, antimicrobial agents, and
hematopoietic cell growth stimulating agents. For instance, in a recent study of ET-18-OCH3 and a liposomal incorporated
ET-18-OCH3, it was found that apoptosis is triggered by this ether lipid by
induction of caspase activation through the release of cytochrome c in a Bcl-XL -
sensitive manner but independently of the CD95 (APO-1/Fas) ligand/receptor
system.27"28 CD95 is a surface membrane molecule involved in cell activation and
apoptosis. It is expressed by a variety of hematopoietic cells, such as
CD34+/CD38+ stem cells, myeloid cells and lymphocytes. Accordingly, the
compounds according to the invention particularly when formulated in a liposome,
may be used as an adjunct for the treatment of tumors in combination to
myelosuppressive chemo-therapeutic drugs and/or those that use the
CD95-ligand/receptor system to trigger apoptosis.
As noted above, the compounds administered to a patient are in the form of
pharmaceutical compositions described above. These compositions may be
sterilized by conventional sterilization techniques, or may be sterile filtered. When
aqueous solutions are employed, these may be packaged for use as is, or
lyophilized, the lyophilized preparation being combined with a sterile aqueous
carrier prior to administration. The pH of the compound preparations typically
will be between 3 and 11, more preferably from 5-9 and most preferably from 7
and 8. It will be understood that use of certain of the foregoing excipients,
carriers, or stabilizers will result in the formation of pharmaceutical salts. As mentioned above, in a preferred embodiment, the compounds according
to the invention will not aggregate platelets. With respect to avoiding platelet
aggregation, various structural modifications to PAF have been studied, which
provide guidelines for modifications that can be made to the antineoplastic ether
lipids. For instance, PAF activity requires an ether linkage at the sn-1 position.
Interestingly, unlike PAF, compounds having a sulfonate or sulfamoyl linkage at
the sn-2 position, may not be susceptible to PLA2 hydrolysis.33 When the ether
linkage is replaced with an ester linkage, the compound becomes susceptible to
PLA inactivation, and no platelet aggregation is observed. It is thus likely that
such compounds may survive enzymatic hydrolysis conditions in aiding prolong
circulation and perhaps could yield potent and selective anti-neoplastic effects.
Further, the D isomer generally elicits no platelet aggregation. Additionally,
when the acetyl group occupying the sn-2 position in PAF is replaced with another
group, PAF activity may be decreased. In this regard, it was found that although
replacement with propionyl doesn't decrease the activity, for every additional
methylene unit added, the activity drops 10 fold compared to PAF. Additionally,
when the acetyl group occupying the sn-2 position in PAF is replaced with a
hydroxyl group (as in lyso PC shown in Figure 1), there is no PAF activity.
While not wishing to be bound by theory, the lack of PAF activity may be due to
the susceptibility of the hydroxyl group to acylation. Finally, when the choline headgroup in PAF is replaced with another moiety, the platelet aggregation effect
is diminished or non-existent.
The L isomer of ET-18-OCH3 elicits a platelet aggregation response in dog
whole blood, most likely due to its structural similarity to PAF. This response
likely reflects an inherent promiscuity in the PAF receptor for dogs. This
response can be blocked by PAF antagonists. Although no platelet aggregation
has been observed using human blood from healthy volunteers, an aggregation
event has been noted in platelet rich plasma (PRP) processed from the blood of
healthy individuals, and in the whole blood of a few cancer patients. The
physiochemical changes responsible for this have not yet been defined.
One way to avoid any potential hematological problems with ET-18-OCH3 or
other ether lipids has been to replace the L isomer with the D isomer, which
doesn't elicit an aggregation response in dog whole blood. Particularly in cases
where the D isomer exhibits roughly the same activity as the L isomer in terms of
cytotoxicity and hemolytic activity, the D isomer is a likely candidate if a
replacement is desired.
In a preferred embodiment of the invention, one or more of these factors are
taken into account in order to produce a compound that does not exhibit a platelet
aggregation effect.
In a further embodiment of the invention, the compounds will also not lyse
red blood cells. If however, the compounds do lyse red blood cells, it is often possible to use a liposome carrier to minimize this effect. For instance, although
ET-18-OCH3 has exhibited antitumor activity in several animal tumor models,8 its
clinical use has been restricted by systemic toxic effects, e.g. hemolysis. In this
regard, a stable liposomal system was developed that would incorporate ET-18-
OCH3 into the bilayer such that its release (exchange out) would be reduced.
Using -molecular monolayer studies of shape complementarity and formulation
optimization, a lipid system which attained ideal packing between the host lipids
and ET-18-OCH3, resulting in a minimized hemolytic activity was determined.
This a system, known as ELL-12, is now being evaluated in clinical trials,23
Using such system, hemolysis has not presented a problem at doses exceeding
those previously tried for the free compound.
In yet another embodiment of the invention, the antineoplastic ether lipids
have desirable pharmacokinetic properties. For instance, it may be desirable to
use a compound that is resistant to "rapid metabolism. " While not wishing to be
limited by theory, lyso PC as shown in Figure 1, is thought to be short lived
because (1) the ester linkage is susceptible to phospholipase cleavage to produce
GPC and (2) lyso PC's free hydroxyl is susceptible to acyltransferases. In
contrast, ET-18-OCH3 is thought to be resistant to the hydrolysis by
membrane-associated phospholipases Al and A2 (PLAl and PLA2) due to its ether
linkages with the sn-1 C 18 chain and sn-2 methyl group. Further, the choline and phosphocholine moieties are known targets for
phospholipases C and D hydrolysis, which yields alkyl-glycerol and
phosphocholine or phosphatidic acid and choline, respectively. One recent
investigation, has shown that ET-18-OCH3 at and above its cytotoxic
concentrations did not inhibit phosphocholine-specific phospholipase C and
phospholipase D, suggesting that ET-18-OCH3 is not their primary target and
could survive in biological membranes.24 However, other studies revealed that
ET-18-OCH3 hexadecylphosphocholme (HPC) can be metabolized by PL-D, thus
making an argument to replace the choline moiety to avoid PC specific PL-D
hydrolysis.25"26
Likewise, phosphonocholine ET-18-OCH3 analogs having a methylene
residue instead of oxygen between the phosphorus and the glycerol moiety, could
significantly help in providing less susceptibility to PL-C Furthermore,
modifying the headgroups with entities bulkier than choline may reduce
susceptibility to choline-specific PL-D as well. This inaccessibility to
phospholipases may allow these compounds to behave as long-acting
anti-neoplastic agents.
In an embodiment of the invention, one or more of these factors are taken
into account to produce novel ether lipid compounds that are stable to potential
phospholipase degradation. Specific embodiments of the invention will now be described through
examples. The following synthetic and biological examples are offered to
illustrate this invention and are not to be construed in any way as limiting the
scope of this invention.
EXAMPLES
In the examples below, the following abbreviations have the following
meanings. If an abbreviation is not defined, it has its generally accepted meaning.
bd = broad doublet
bs = broad singlet
c = concentration
d = doublet
dd = doublet of doublets
ddd = doublet of doublets of doublets
DMF = dimethylformamide
DMSO = dimethyl sulfoxide
g — grams
hept. = heptuplet
J = coupling constant
m = multiplet M molar
max — maximum
mg = milligram
min. = minutes
mL = milliliter
mM = millimolar
mmol = millimole
N = normal
ng = nanogram
nm = nanometers
OD or o.d. = optical density
q = quartet
s = singlet
sept = septuplet
t = triplet
THF = tetrahydrofuran
tic = thin layer chromatography
μL = microliter
The antibodies were obtained from the following vendors: Transduction
Laboratories, Lexington, KY (Raf-1, PKB/AKT); New England Biolabs Inc, Beverly, MA (phospho-MAP kinase and phospho- PKB/AKT); Santa Cruz Inc,
Santa Cruz, CA (ERK-1, ERK-2); fetal bovine serum (FBS) from Hyaclone
(Logan, UT).
Additionally, the term "Aldrich" indicates that the compound or reagent used
in the following procedures is commercially available from Aldrich Chemical
Company, Inc. , 1001 West Saint Paul Avenue, Milwaukee, WI 53233 USA; the
term "Fluka" indicates the compound or reagent is commercially available from
Fluka Chemical Corp., 980 South 2nd Street, Ronkonkoma, NY 11779 USA; the
term "Lancaster" indicates the compound or reagent is commercially available
from Lancaster Synthesis, Inc., P.O. Box 100, Windham, NH 03087 USA; and
the term "Sigma" indicates the compound or reagent is commercially available
from Sigma, P.O. Box 14508, St. Louis, MO 63178 USA.
Unless otherwise stated, all temperatures are in degrees Celsius.
NMR spectra were recorded on an IBM-Bruker 200-MHZ or a Bruker 400-
MHZ Spectrometer with Me4Si as internal standard. Infrared spectra were
recorded on a Perkin-Elmer 1600 FT spectrophotometer. Optical rotations were
measured on a JASCO Model DIP- 140 digital polarimeter using a 1-dm cell.
Methylene chloride and pyridine were distilled from calcium hydride and barium
oxide, respectively. Chloroform was distilled from P2O5. All other synthetic
reagents were used as received unless otherwise stated. In these synthetic methods, the starting materials can contain a chiral center
and, when a racemic starting material is employed, the resulting product is a
mixture of R,S enantiomers. Alternatively, a chiral isomer of the starting material
can be employed and, if the reaction protocol employed does not racemize this
starting material, a chiral product is obtained. Such reaction protocols can involve
inversion of the chiral center during synthesis. Alternatively, chiral products can
be obtained via purification techniques which separates enantiomers from a R, S
mixture to provide for one or the other stereoisomer. Such techniques are well
known in the art.
PART I: PREPARATION OF COMPOUNDS
The compounds of the invention were synthesized with high percentage
enantiomeric excess starting from the commercially available optically active O-
benzylglycidyl ether precursor.
Routes to Synthesis of Ether Lipids
Figure imgf000047_0001
A
CH3S02CI
(OCH2CH2)n)OCH3 H=CH(CH2)7CH3 (trans) H=CH(CH2)7CH3 (cis)
Figure imgf000047_0002
(R)-O-Benzyl glycidyl ether A was subjected to the oxirane ring opening by
the appropriate alcohol in presence of catalytic amounts of boron triflate etherate
to yield B. Compound B was then reacted with methanesulfonyl chloride in
methylene chloride and pyridine to yield C. Cleavage of the benzyl group of
compound C was carried out by hydrogenolysis on 5 % Pd on carbon under hydrogen gas to yield compound D. Finally, compound D was phosphorylated
with phosphorus oxychloride and friemylamine at ambient temperature followed by
coupling with choline tosylate/pyridine and then hydrolysis to yield compounds 1,
2 and 3.
PART II: METHODS FOR EVALUATION OF COMPOUNDS
The ether lipid compounds according to the invention may be screened by
any acceptable method(s) used in the field. For example, the ether lipid
compounds may be examined with respect to the ability of the new compounds (1)
to aggregate platelets (i.e. , mimic PAF), a specific toxicity to avoid or minimize,
(2) to lyse red blood cells, a non-specific toxicity for which a liposome carrier may
be needed, (3) to inhibit growth of tumor cells as a measure of activity, and (4) to
inhibit growth of normal cell lines as compared to tumor cells, a measure of
selectivity. Representative screening assays are contained in the following
description.
After using these screening assays, candidates deemed suitable for in vivo
testing were then synthesized in large scale quantities and some of this material
was sent to the NCI's Developmental Therapeutics Program for a battery of
growth inhibitory studies against various human tumor cell lines (60 cell lines, 9
different panels). The remainder of the material was used to assess in vivo
efficacy using murine tumor models. Additionally, studies were conducted on the lead candidates to discern
mechanism of action with particular emphasis on these agents apoptotic ability as
measured by caspase 3 activity. The details (materials and methods) for these
various assays are described further in the following discussion.
I. Platelet Aggregation Assay
Platelet aggregation in whole blood is measured using a whole blood
aggregometer from Chronolog Corp. The species most often used is dog, since
this species has consistently shown a strong platelet aggregation response in whole
blood to L-ET-I8-OCH3. However, any species, including human may be used.
Briefly, whole citrated blood is diluted 1:2 with sterile saline and placed in a
warm chamber with a mini stir bar. An electronic probe, measuring electrical
resistance, is inserted in the sample. The aggregometer is calibrated and the
baseline is observed to detect any spontaneous aggregation. The test sample is
added and allowed to run for at least 6 minutes. If the sample is an agonist the
platelets will start to aggregate and stick to the electronic probe causing resistance
across the electrodes to increase. This resistance, in ohms, is measured 6 minutes
post addition of sample. The test samples are run at 25, 100, 200, 400 and 800
uM and compared to 100 uM L-ET-18-OCH3. A. Collection of Blood Sample for in vitro Platelet Aggregation Testing
Platelet aggregation was assessed using dog whole blood, a system found to
be highly sensitive as it has been demonstrated that the L-isomer of ET-18-OCH3
invokes aggregation at relatively modest concentrations (but not the D-isomer).
Venous blood is collected in 4.5 mL Vacutainer tubes containing 0.129 M
sodium citrate using a 21G needle or larger. Blood is immediately mixed by gentle
inversion 15-20 times and kept at room temperature. Dilution ratio is 1 to 9 (3.8%
citrate solutiomblood). One Vacutainer tube (2.0 mL) containing EDTA is also
collected for platelet counts. Complete blood counts (CBC) are measured on the
CDC Technologies Hemavet 1500 to ensure that the test subject's platelets are
within normal range. Any vials of hemolytic blood or blood containing any clots
should be discarded. Platelet aggregation testing must be completed within 3
hours of blood collection. After this time the ability of platelets to aggregate
decreases.
B. Procedure for dilutions
Test samples are provided either as powder or solutions. Cloudy solutions
were warmed to —50° C to dissolve any particles. Dilutions were prepared in PBS
or saline at 40X concentration. (25 uL test sample are added to 1000 uL diluted
blood (1:40)). All compounds were diluted in saline. However, if samples were poorly
soluble, they may be diluted in DMSO. It should be noted that since DMSO itself
may elicit a response, a control for DMSO should also be used.
C Procedure for in vitro Platelet Aggregation Test using CHRONO-LOG
Aggregometer Model 560-CA-
The following protocol was used:
1. Turn on aggregometer, aggrolink, monitor, computer and printer at least
15 minutes before testing to warm aggregometer to 37 °C
2. Double click on "AGGLINK" in Windows.
3. Set stir speed to 1000 for whole blood.
4. Set up small beaker with deionized water to clean impedance probes after
each sample. Set up 2 plastic cuvettes with — ImL of saline to store probes in
warming chambers between tests. Set up 1 small plastic cuvette or test tube with
— 2 mL of saline to clean pipet after addition of test articles.
5. Click on "aggregometer"
Click on "test procedure" and set parameters
Procedure Name (Ether Lipid platelet aggregation test)
Channels = 4
Duration = 6:00 (min: sec)
Reagent (test article or L-EL control) Concentration (25-800 uM)
Stirrer = 1000
Gain = 20/5 (20 ohms = 5 blocks)
Enter OK to exit
6. Click on "aggregometer"
Click on "run test" and set parameters
Enter Subject Information
Last Name = WB 1:2
First Name: Subject
ID#= time
Hospital = N/A
Test Procedure (Edit if necessary)
Enter OK to exit
Make sure aggregometer temperature reads 37 °C before testing.
7. Place 1 mL plastic cuvettes into warming wells.
8. Add 1 disposable siliconized stir bar to each cuvette.
9. Add 500 uL saline to 2 cuvettes
10. Add 500 uL whole titrated blood to the two saline containing cuvettes.
M-1000 positive displacement pipet is recommended to measure blood volume.
11. Warm diluted blood 5 min at 37° C in warming wells. 12. Transfer diluted blood samples (in duplicate) to aggregation chambers
and insert impedance probes. Close doors.
13. Calibrate each chamber (This must be done for each test run). Zero
channels to baseline with zero knob. Hold in calibration button and adjust gain to
50%. Observe steady baseline for 1 minute. Recalibrate if necessary.
14. Open chamber door and add 25 uL test sample to each cuvette
15. Rinse capillary pipet piston with saline after each use.
16. Allow test to run at least 6 minutes.
17. Click "aggregometer" and then click on "stop test"
18. Click "Edit"
19. Click "set start & stop time. " Select channel 1 and hold down both
mouse buttons while moving vertical start line to 3-5 seconds prior to sample
injection (the stop time will automatically move to 6 min after start time).
20. Click done. Repeat step 15 and 16 to select channel 3.
21. Click "Edit".
22. Click "compute slope & amplitude" and then check that both channels
are set for 6 min run. Click OK. The aggregometer automatically calculates the
ohms amplitude.
23. Click "file" and click "Save"
24. Click "File"
25. Click "close" 26. Remove impedance probe and place in beaker of deionized water
27. Gently remove any aggregated platelets from probe and place into warm
saline prior to next test.
28. Discard test sample in biological waste.
29. Print out files Platelet Aggregation.
P. Results
Table 1: Platelet aggregation of New Ether Lipid Analogs.
Compound Relativ e Platelet A ggregation (Ohms) Platelet aggregation
25 μM 100 .M 200 μM 400 μM 800 μM
L-ET-I8-OCH3 13 14 12 nd nd + + +
D-ET-I8-OCH3 0 0 0 nd nd -
1(R) 0 0 0 0 0 -
KS) 0 0 0 0 0 -
Platelet aggregation in dog whole blood was measured in Ohms. For all experiments, O.lμM PAF and 100 μM ET-18-OCH3 were included as positive controls.
As shown by the data in Table 1, neither compound 1(R) nor 1(S) exhibited
a platelet aggregation response.
While not wishing to be bound by theory, steric hindrance at the sn-2
position may be the reason why the sulfonate compounds 1(R) and 1(S) do not
evoke a platelet aggregation.
Other testing showed the substitution of phosphorus for nitrogen or the
introduction of a ring system in the headgroup chain did not prevent recognition
of these compounds by the PAF receptor; in both instances the number of
methylenes in the headgroup chain were 2. It was also found that when the number of methylenes in the headgroup was
increased or the positive charge was removed, no aggregation was noted indicating
that those changes sufficiently altered binding to the PAF receptor.
II. In Vitro Hemolytic Activity Assay
The hemolytic activity of the various compounds was compared to ET-18-
OCH3. A compound that is more hemolytic than ET-18-OCH3 might require a liposome to minimize such side effects, while a compound that is equal or less hemolytic may not require a liposomal carrier (at least under the conditions established here for this screening study).
A. Hemolysis Assay with Washed Human Red Blood Cells
Venous blood was collected in 10 mL Vacutainer tubes containing EDTA
using a 21G needle or larger. Blood was immediately mixed by gentle inversion
15-20 times and kept at room temperature. The blood was transferred from 1
EDTA tube to a 50 mL conical tissue culture tube and the volume was brought up
to 50 mL with PBS.
The blood was centrifuged for 10 minutes at 1500 RPM. The supernatant
was removed and the blood was resuspended up to 50 mL with PBS. Next the
blood was centrifuged for 10 minutes at 1500 RPM. The supernatant was removed, and 2.0 mL of packed red blood cells were
carefully transferred, using positive displacement pipet, into a fresh conical tissue
culture tube. Next, 48 mL PBS was added to achieve a 4% washed RBC solution.
Next, 25, 50, 100 and 200 uM stock solutions of test sample in phosphate
buffered saline (PBS) as follows:
Stock Solution Test Sample PBS
200 uM 200 uL of 20 mM + 19.8 mL
100 uM 5 mL of 200 uM + 5 mL
50 uM I mL of 100 uM + I mL
25 uM 500 uL of 50 uM + 500 uL
Next, 0.5 mL of 4% washed RBC was added to 0.5 mL test sample dilutions
(in duplicate). The final concentration of test sample was 50% of working stock
solution.
The samples were capped or sealed with Parafilm and the samples were
gently mixed. The blood was incubated at 37 °C in gentle rotating water bath for
30 minutes. The samples were centrifuged for 10 minutes at 1500 RPM. Next,
200 uL of supernatant was transferred to a cuvette and 1 mL deionized water was
added. Absorbance was measured at 550 nm vs. a water blank. Next, H10 and H50
were determined by graphing "Absorbance" vs. "Test Sample Concentration. " B. Hemolysis Assay with Whole Human Blood
Venous blood was collected in 10 mL Vacutainer tubes containing EDTA
using a 21G needle or larger. The blood was immediately mixed by gentle
inversion 15-20 times and kept at room temperature.
Stock solutions of 20 mM test sample in phosphate buffered saline (PBS)
were prepared as follows:
Working Stock Test Sample PBS
20,000 uM 100 uL of 20,000 uM
10,000 uM 100 uL of 20,000 uM + 100 uL
5,000 uM 100 uL of 10,000 uM + 100 uL
2,500 uM 100 uL of 5,000 uM + 100 uL
1,000 uM 100 uL of 2,500 uM + 100 uL
500 uM 100 uL of 1,000 uM + 100 uL
Then 270 uL of whole blood was aliquotted in duplicate mini test tubes using
positive displacement pipette. Next, 30 uL of each working stock solution, in
duplicate, was added to the whole blood. Next, 30 uL of PBS was added for
background control. The samples were capped or sealed with Parafilm and gently
mixed. The blood was incubated at 37° C in gentle rotating water bath for 30
minutes. Final concentration of test sample was 10% of working stock solution. Total hemolysis samples of 1 % and 10% whole blood samples in deionized
water were prepared as follows:
1: 100= 10 uL whole blood + 990 uL deionized water
1: 10= 100 uL whole blood + 900 uL deionized water
The samples were freeze thawed 3X in liquid nitrogen then water bath. The
samples were then centrifuged 10 minutes at 1500 RPM. Next, 100 uL of
supernatant was transferred to a cuvette and 1 mL deionized water was added.
The absorbance was read at 550 nm vs. water blank.
Hj0 and H50 were calculated by graphing Percent Total Hemolysis vs. Test
Sample Concentration. The Percent Total Hemolysis = (average o.d of test
sample)/(average o.d. of total hemolysis sample) X 100
Hemolytic activities for L-ET-18-OCH3 and the corresponding liposomal
formulation, L-ELL-12 are shown in Table 2. As shown by the data in Table 2,
inclusion of compounds in liposomes dramatically decreased hemolytic activity.
Table 2. Hemolytic Activity
Washed Human RBCs Whole Human Blood H,„ (βM) Hm (μM. H,n (μ ) H^ faM)
Free Compound
L-ET-I8-OCH3 11, 10.5 17, 15 600, 550, 700 2000, > 2000
Liposome Systems L-ELL-12 350 >2000 > 2000 > >2000
H10 and H50 values are the concentrations at which the ether lipids produce 10% or 50% hemolysis, respectively. D-ET-18-OCH3 historically produced the same values as the L isomer and is not shown here. Some experiments were repeated thus the additional entries. For the new liposome formulations, all liposomes were extruded to approximately 100 nm in mean diameter. Choi = cholesterol; DOPC = dioleoylphosphatidylcholine; DOPE-GA = dioleoylphosphatidylemanolarnine- glutaric acid (glutaric acid is covalently attached via the headgroup nitrogen ); CHS = cholesteryl- hernmisuccinate.
TTI. In Vitro Growth Inhibition (GTr0) Assay
A. Cell line maintenance
The following cell lines were selected from the cell bank for screening and
GI50 studies: MCF-7: human breast tumor, MCF-7/ADR: MCF-7 adriamycin
resistant subline, HT-29: human colon carcinoma, A-549: human non-small cell
lung cancer, NIH-3T3: mouse swiss embryo fibroblast and WI-38: human lung
fibroblast, SKMEL-28: human melanoma, Lewis Lung: mouse lung carcinoma,
DU-145: prostate carcinoma, B16F10: mouse melanoma, L1210: murine
lymphocytic leukemia, P-388: murine leukemia, U-937: human histolytic
lymphoma. Except HT-29, WI-38, NIH-3T3 and Lewis Lung which were
obtained from the American Type Culture Collection (Rockville, MD) all the other
cell lines were obtained from National Cancer Institute - Frederick Research
Facility (Frederick , MD). All the cell lines were grown in RPMI-1640 medium containing 10% fetal bovine serum (FBS) except WI-38 and DU-145 which were
grown in EMEM + 10% FBS at 37°C, 5% CO2 and 100% humidity and NIH-
3T3, Lewis Lung, B16F10 and L1210 which were grown in DMEM containing
10% FBS (10% HS for L1210). All the adherent cell lines were detached from the
culture flasks by addition of 2-3 ml of 0,05% trypsin-EDTA. Thereafter, trypsin
was inactivated by addition of lOmL of 10% serum-containing RPMI-1640
medium. Cells were separated into a single-cell suspension by gentle pipetting
action. Depending on the cell type, 3,000 to 10,000 cells were plated onto 96-well
plates a day prior to the drug treatment, in a volume of 100 μl per well.
B. Drug Treatment
The test compounds were dissolved in PBS or saline at a stock concentration
of approximately 20 mM, which is 400 times the desired final maximum test
concentration. The stock solutions were then diluted with complete medium to
twice the desired final concentration. A 100 μl aliquot of each dilution was then
added to the designated wells. After 3 days of incubation, cell growth was
determined.
C Sulforhodamine B (SRB) assay
The SRB assay was performed with minor modifications to the method
described by Monks, A., Scudiero, D., Skehan, P., Shoemaker, R., Paull, K., Vistica, D.,Hose, C, Langley ., Cronise,P., and Vaigro-Wolff, A., "Feasibility
of a high-flux anticancer drug screen using a diverse panel of cultured human
tumor cell lines, " J Natl Cancer Inst. 83: 757-766 (1991), which is hereby
incorporated by reference in its entirety. Following drug treatment, cells were
fixed with 50μl of cold 50% (wt/vol) trichloroacetic acid (TCA) for 60 minutes at
4°C. The supernatant was discarded, and the plates were washed six times with
deionized water and then air dried. The precipitate was stained with 100/χl SRB
solution (0.4% wt/vol in 1 % acetic acid) for 10 minutes at room temperature, and
free SRB was removed by washing three times with 1 % acetic acid, and the plates
were then air dried. Bound SRB was solubilized with Tris buffer (lOmM), and the
ODs were read using an automated plate reader (Bio-Rad, Model 3550-UV) at
490nm. Background values were subtracted from the test data, and the data was
calculated as a % of control. The GI50 represents the concentration of test agent
resulting in 50% of net growth compared to that of the untreated samples. In this
assay, ODs were also taken at time 0 ( the time the drugs were added ) If the ODs
of the tested samples were less than that of time 0, cell death had occurred.
Percentage growth was calculated by the method described by Peters, A.C,
Ahmad, I., Janoff, A.S., Pushkareva, M. Y., and Mayhew, E. "Growth Inhibitory
effects of liposome-associated l-o-octadecyl-2-o-methyl-sn-glycero-3-
phosphocholine, " Lipids. 32:1045-1054 (1997). The raw optical density data was
imported into an Excel spreadsheet to determine dose responses. Percentage growth was calculated as follows: (T-T0)/(C- T0)x 100 where (T)=mean optical
density of treated wells at a given drug concentration, (T0)=mean optical density
of time zero wells, and (C)=mean optical density of control wells, or if T < T0
where cell killing has occurred, then percent death can be calculated as follows:
(T- T0)/ (T0) x 100. By varying drag concentration, dose response curves were
generated and the GI50 values were calculated. The GI50 values for each experiment
were calculated using data obtained from three duplicate wells on two separate
plates. The mean GI50's from each independent experiment.
D. Cell Growth Assay
To determine the growth inhibition in the suspension cell lines, cell numbers
were directly counted instead of using the SRB assay which determines the total
cell protein. One day prior to the drug treatment, 40,000 cells per well were
seeded into 24-well plates in a volume of 0.5mL. Stock solutions were diluted
with complete medium to twice the desired final concentrations, and then 0.5mL
aliquots of each dilution were added to the designated wells. After 3 days
incubation, cell growth was determined by counting cell number using a coulter
counter (Z-M, coulter). Cell counts were also taken at time 0 and subtracted from
the test results to give net growth. The GI-50 represents the concentration of test
agent resulting in 50 % of net growth compared to that of the untreated control
samples. E. Results
For growth inhibitory evaluation, five human tumor cell lines were used
(U937; HT29; A549; MCF7; MCF7/ADR) and two normal fibroblast cell lines
(NIH-3T3, murine; WI-38, human). For comparison, the activity of free L-ET-
18-OCH3 and D-ET-18-OCH3 was examined.
Table 3: Growth Inliibition of Tumor/Normal Cells
Figure imgf000063_0001
As shown in Table 3, both L and D isomers of ET-18-OCH3 gave essentially identical results with the order of sensitivity for the cells lines being U937>HT29>A549>MCF7>MCF7/ADR, H-3T3 (normal cell line). WI-38 cells were moderately sensitive to both ether lipids with GI50 values of 10-13 μM, which was 3-4 times lower than that for the NIH-3T3 cells at 41-47 μM.
TV. Measurement of DEVDase Activity
In a recent study, it was found that ELL- 12 triggers apoptosis by induction of caspase activation through the release of cytochrome c in a Bcl-xL - sensitive manner but independently of the CD95 (APO- 1/Fas) ligand/receptor system.27 To determine whether the new ether lipids produce their growth inhibitory effects via the same apoptotic mechanism as ELL-12, and to correlate these results with activity, DEVDase activity, which is a specific correlate of Caspase 3 activity, was examined.
Suspension cells were seeded at density 3.2xl05 cells per mL in RPMI-1640 medium (Bio-Whittaker) supplemented with 10% heat inactivated FBS (Bio- Whittaker). Cells were pre-incubated overnight prior to treatment with the ether lipid compounds of the invention. At treatment time cell density was 5xl05 cells per mL. Cells were incubated with the ether lipid compounds of the invention for various periods of time, usually 6 hours. Cells were collected, washed with IxDPBS and resuspended in 110 μl of Buffer A (10 mM HEPES-KOH, pH 7.4, 2 mM EDTA, 0.1 % CHAPS, 5 mM DTT, 1:100 dilution of protease inhibitors cocktail (Sigma)-DTT and protease inhibitors should be added just before use). After 10 min incubation on ice in order to lyse cells, samples were frozen on dry ice/ethanol and kept at -20°C until analysis.
At the time of analysis of DEVDase activity frozen pellets were kept on ice until thawed and after vigorous vortexing samples were centrifuged at 14,000 rpm for 6 min. Supernatant was transferred into another tube and 2 μl, in triplicates, were used for protein measurement using Bradford reagent (Bio-Rad).
Measurement of DEVDase activity was carried out in 100 μl volume, where 10 μg of protein was delivered in 50 μl of Buffer A and 40 μM of substrate Ac- DEVD-AMC was also delivered in 50 μl of Buffer A. All measurements were done in triplicates. Reaction was carried out for 1 hour and generation of fluorescent product of reaction (aminomethylcoumarin) was measured by reading fluorescence at lem 460 nm (lex 360 nm). Changes in DEVDase activity were calculated after subtraction of background fluorescence of substrate incubated without proteins, and were expressed as percent of control (DEVDase activity in untreated cells). Average of DEVDase activity, calculated from few independent experiments, was used to calculate percent of L-ether lipid-induced DEVDase activity.
First, various conditions to compare free ET-18-OCH3 and ELL-12 were examined. As shown in Figure 4, there is indeed a difference between liposomal compound and free drug. This difference is likely due to availability and, consistent with this notion, subsequent experiments showed that longer incubation times caused the differences to diminish (data not shown). However, the 6 hour time frame made for convenient testing and was used for comparative studies.
Table 4. DEVDase Activity of NELs in Jurkat T Cells at 6 Hours
% L-ET180CH3 (DEVDase, 6hrs) Tether lipidl. μM
Lipid 1Q 2Q 2Q N
L-ET-I8-OCH3 100 ± 8.8 100 ± 10.4 100 ± 9.3 7
D-ET-I8-OCH3 79.3 + 9.7 89.2 ± 12.2 92.1 ± 11.8 4
L-ELL-12 66.1 ± 8.1 78.5 + 10.8 71.3 ± 9.4 4
D-ELL-12 56.5 ± 6.8 78.2 ± 10.5 85.5 + 10.4 4
N is the number of replicate experiments. V. In Vivo Toxicity and Therapeutic Methods
A. Toxicity
Intravenous (xl) or Oral (xl)
CDF1 mice (3 /group) were administered a single intravenous or oral dose of the ether lipid compounds to be tested. Mortality was recorded daily and body weights were recorded at least twice weekly for an observation period of 30 days.
B. Therapeutic
B 1 /F10 Murine Melanoma (iv, / iv.)
Female C57/BL6 mice (5/group) were inoculated iv. with 5 x 104 cells in 0.2 L PBS (day 0). On days 10, 12, 14, 16, & 18 post-tumor inoculation, mice were treated iv. with the ether lipid compounds to be tested, along with ELL- 12 (L), D- EL, L-EL or Control (0.9% NaCl). Mice were sacrificed by carbon dioxide inhalation on day 22, lungs were excised, inflated and fixed with 10% Formalin. Lungs were counted "blind" for tumor nodules using a magnifier. The mean number of nodules per treatment group was determined. P388 Murine Leukemia (ipJ iv.)
Female CDF1 mice (7-8/group) were inoculated ip. with 1 x 105 P388 cells in 0.5 mL PBS (day 0). Treatments were administered iv. on days 1, 3, 5, 7, & 9 post inoculation with with the ether lipid compounds to be tested, along with ELL- 12 (L), L-EL, or Control. Mice were checked daily for mortality and the percent of survival was determined.
P388 Murine Leukemia (ip./ ip.)
Female CDF1 mice (4-8/group) were inoculated ip. with 1 x 105 P388 cells in 0.5 mL PBS (day 0). Treatments were administered ip. on days 1 - 10 post inoculation with with the ether lipid compounds to be tested, along with ELL- 12 (L), or Control or on days 1 - 8 post inoculation withNEL-AlS, -A3S, -A3R, -A5S, - A7R, -A9S, -A21S, -A26R, D-EL, L-EL or Control (NaCl). Mice were checked daily for mortality and the percent survival was determined.
LI 210 Murine Leukemia (ip./ iv.)
Female DBA/2 mice (3-5/group) were inoculated ip. with 1 x 105 cells in 0.5 mL PBS (day 0). Treatments were administered iv. on days 1, 3, 5, 7, & 9 post inoculation with with the ether lipid compounds to be tested, along with D-EL, L-EL or Control (NaCl). Mice were checked daily for mortality and the percent survival was determined. DTT145 Human Prostate (so. / iv.)
Male SOD mice (5/group) were inoculated sc. with 2 x 106 cells in O.lmL PBS (day 0) and the tumors were allowed to reach a volume of -250 mm3 at the start of treatment. Treatments were administered iv. on days 27, 28, 29, 30, & 31 with with the ether lipid compounds to be tested, along with ELL- 12 (D), ELL- 12 (L), L- EL, or Control (NaCl). Tumors were measured with calipers and tumor volume
(mm3) was calculated as (Length x (Width)2 x p).
MX-1 Human Mammary (sc. / iv.)
Female SCJD mice (5/group) were inoculated sc. with 10 mg/O.lmL tumor mince (day 0), and the tumors were allowed to reach a volume of ~200mm3 at the start of treatment. Treatments were administered iv. on days 13, 15, 17, 19, & 21 with the ether lipid compounds to be tested, along with ELL- 12 (D), ELL- 12 (L), L- EL, or Control (NaCl). Tumors were measured with calipers and tumor volume
(mm3) was calculated as (Length x (Width)2 x p).
The invention has been described with reference to specific embodiments. Substitutions, omissions, additions and deletions may be made without departing from the spirit and scope of the invention defined in the appended claims. From the foregoing description, various modifications and changes in the composition and method will occur to those skilled in the art. All such modifications coming within the scope of the appended claims are intended to be included therein. All of the above publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

Claims

The claimed invention is:
1. An ether lipid having formula (I), or a pharmaceutically acceptable salt, isomer or prodrug thereof:
Figure imgf000070_0001
Formula (I) wherein:
R1 is selected from the group consisting of ~CH2CH2(OCH2CH2)mO—CH3 and --(CH2)nCH=CH(CH2)pCH3;
R2 is selected from the group consisting of —OR3 and
Figure imgf000070_0002
R3 and R4 are each independently selected from the group consisting of C 4 alkyl and H; and
n, m and p are each independently an integer from 1 to 10.
2. An ether lipid of Claim 1 , wherein n, m and p are each independently an integer from 1 to 5.
3. An ether lipid of Claim 2, wherein n, m and p are each independently an integer from 1 to 3.
4. An ether lipid of Claim 1 , wherein:
R2 is selected from the group consisting of — OMe and — OSO2Me.
5. An ether lipid of Claim 4, wherein R2 is — OSO2Me.
6. An ether lipid of Claim 4, wherein R2 is — OMe.
7. An ether lipid of Claim 1, wherein:
R2 is selected from the group consisting of — OEt and — OSO2Et.
8. An ether lipid of Claim 1 , wherein the ether lipid is selected from the group consisting of:
Figure imgf000071_0001
C^
Figure imgf000071_0002
Figure imgf000071_0003
9. An ether lipid of Claim 8, wherein the ether lipid is:
Figure imgf000072_0001
10. An ether lipid of Claim 8, wherein the ether lipid is:
Figure imgf000072_0002
11. An ether lipid of Claim 8, wherein the ether lipid is:
Figure imgf000072_0003
12. An ether lipid of Claim 8, wherein the ether lipid is optically active.
13. An ether lipid of Claim 9, wherein the ether lipid is the D enantiomer.
14. An ether lipid of Claim 10, wherein the ether lipid is the D enantiomer.
15. An ether lipid of Claim 11 , wherein the ether lipid is the D enantiomer.
16. A pharmaceutical composition comprising a pharmaceutically effective amount of an ether lipid of Claim 1 or a pharmaceutically acceptable salt, isomer or prodrug thereof, and a pharmaceutically acceptable carrier.
17. A pharmaceutical composition comprising:
(a) a liposome, emulsion or mixed miscelle carrier and
(b) a pharmaceutically effective amount of an ether lipid of Claim 1 or a pharmaceutically acceptable salt, isomer or prodrug thereof.
18. A liposome comprising an ether lipid of Claim 1 or a pharmaceutically acceptable salt, isomer or prodrug thereof.
19. A method of treating a mammal afflicted with a cancer which comprises administering to the mammal a therapeutically effective amount of the pharmaceutical composition of Claim 16 comprising from about 0.1 mg of the ether lipid per kg of the body weight of the mammal to about 1000 mg per
kg-
20. A method of Claim 19, wherein the cancer is selected from the group consisting of lung cancers, brain cancers, colon cancers, ovarian cancers, breast cancers, leukemias, lymphomas, sarcomas and carcinomas.
21. The method of Claim 19, comprising administering to the mammal an additional biologically active agent.
2. The method of Claim 21, wherein the additional biologically active agent is selected from the group consisting of antineoplastic agents, antimicrobial agents, and hematopoietic cell growth stimulating agents.
PCT/US2004/000359 2003-01-09 2004-01-09 Antineoplastic ether lipid compounds with modifications at the sn-1 carbon WO2004062595A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US43878703P 2003-01-09 2003-01-09
US60/438,787 2003-01-09

Publications (2)

Publication Number Publication Date
WO2004062595A2 true WO2004062595A2 (en) 2004-07-29
WO2004062595A3 WO2004062595A3 (en) 2004-12-09

Family

ID=32713382

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/000359 WO2004062595A2 (en) 2003-01-09 2004-01-09 Antineoplastic ether lipid compounds with modifications at the sn-1 carbon

Country Status (1)

Country Link
WO (1) WO2004062595A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006019907A1 (en) * 2006-04-28 2007-10-31 Müller-Enoch, Dieter, Prof. Dr. Use of substituted glycerin derivative in the preparation of a pharmaceutical composition for the prevention or treatment of e.g. cancer disease, pathological sequence of alcohol abuse, viral hepatitis and toxic nerve disorder

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4562179A (en) * 1982-04-19 1985-12-31 Fujisawa Pharmaceutical Co., Ltd. Phospholipid derivatives, and pharmaceutical composition of the same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4562179A (en) * 1982-04-19 1985-12-31 Fujisawa Pharmaceutical Co., Ltd. Phospholipid derivatives, and pharmaceutical composition of the same

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006019907A1 (en) * 2006-04-28 2007-10-31 Müller-Enoch, Dieter, Prof. Dr. Use of substituted glycerin derivative in the preparation of a pharmaceutical composition for the prevention or treatment of e.g. cancer disease, pathological sequence of alcohol abuse, viral hepatitis and toxic nerve disorder

Also Published As

Publication number Publication date
WO2004062595A3 (en) 2004-12-09

Similar Documents

Publication Publication Date Title
TWI630201B (en) Compounds and uses thereof for the modulation of hemoglobin
US4734225A (en) D-mannite derivatives as starting products for the synthesis of phospholipids
EP2351730B1 (en) Ethoxy diphenyl ethane derivates, preparation processes and uses thereof
US20130190524A1 (en) Cyclitols and their derivatives and their therapeutic applications
CZ541288A3 (en) 4&#39;-phosphate epipodophyllotoxin glucosides, process of their preparation and pharmaceutical preparation in which they are comprised
JP2002500227A (en) Combretastatin A-4 prodrug and its trans isomer
PL204458B1 (en) Phosphoramidate, and mono-, di-, and tri-phosphate esters of (1r, cis)-4-(6-amino-9h-purin-9-yl)-2-cyclopentene-1-methanol as antiviral agents
JP2009502986A (en) Elianin salt, method for preparing the same, and drug composition containing the same
EP0061872B1 (en) Ethyleneglycol derivatives, their production and use
EP1305322B1 (en) Derivatives of branched-chain lipophilic molecules and uses thereof
US4710579A (en) 2-(acetoacetyloxy)-3-(octadecyloxy)propyl-3-trimethylammoniopropyl phosphate or a pharmaceutically acceptable salt thereof
IL155619A (en) Glycerophosphoinositol derivatives as modulators of cytosolic a2 phospholipase
EP2727927B1 (en) Camptothecin derivative, method for preparing same, pharmaceutical composition and use thereof
EP1574216B1 (en) Inhibitors of phosphatidyl Myo-Inositol Cycle
US20030008850A1 (en) Phosphoramide compounds
WO2004062595A2 (en) Antineoplastic ether lipid compounds with modifications at the sn-1 carbon
US20060116354A1 (en) Antineoplastic ether lipid compounds with modifications at the sn-3 carbon
US20060122153A1 (en) Antineoplastic ether lipid compounds with modifications at the sn-2 carbon
WO2004062586A2 (en) Antineoplastic ether lipid compounds
WO2004062596A2 (en) Antineoplastic ether lipid compounds having a four carbon backbone
FI109699B (en) Process for the preparation of pharmacologically active liponucleotides of seconucleosides
US8097752B2 (en) Antiprotozoal ring-substituted phospholipids
US7323601B2 (en) Enanitomers of unsaturated alkyllysophosphonocholines and use as anti-neoplastics
AU2002244079A1 (en) Enantiomers of unsaturated alkyllysophosphonocholines and use as anti-neoplastics
NO328854B1 (en) Bis- (N &#39;, N&#39;-bis- (2-haloethyl) amino) phosphoramidate, pharmaceutical composition comprising the compound, method of preparation and use of the compound.

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase