EP1292562A1 - Triterpenoid derivatives - Google Patents

Triterpenoid derivatives

Info

Publication number
EP1292562A1
EP1292562A1 EP01936618A EP01936618A EP1292562A1 EP 1292562 A1 EP1292562 A1 EP 1292562A1 EP 01936618 A EP01936618 A EP 01936618A EP 01936618 A EP01936618 A EP 01936618A EP 1292562 A1 EP1292562 A1 EP 1292562A1
Authority
EP
European Patent Office
Prior art keywords
compound
formula
methyl
hal
single bond
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.)
Withdrawn
Application number
EP01936618A
Other languages
German (de)
French (fr)
Inventor
Marian Hajduch
Jan Sarek
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.)
Karlova Univerzita v Praze
Univerzita Palackeho V Olomouci
Original Assignee
Karlova Univerzita v Praze
Univerzita Palackeho V Olomouci
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 GBGB0012528.6A external-priority patent/GB0012528D0/en
Priority claimed from GB0012823A external-priority patent/GB2362649A/en
Application filed by Karlova Univerzita v Praze, Univerzita Palackeho V Olomouci filed Critical Karlova Univerzita v Praze
Publication of EP1292562A1 publication Critical patent/EP1292562A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • C07J63/008Expansion of ring D by one atom, e.g. D homo steroids

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to the use of a compound of formula (I), or a pharmaceutically acceptable salt, crystal form, complex, hydrate, or hydrolysable ester thereof, in the preparation of a medicament for treating a patient suffering from leukaemia, cancer or other proliferative disorder. A further embodiment relates to the use a compound of formula (I) in an assay for detecting the phosphorylation state of cellular substrates. The present invention also relates to novel compounds of formula (I), and the chemical synthesis thereof.

Description

TRΠΈRPENQID DERIVATIVES
The present invention relates to the therapeutic use and the biological activity of triterpenoid derivatives. The invention further relates to novel triterpenoid derivatives.
To date, the prior art has primarily focussed on compounds that are capable of regulating the cell cycle by virtue of inhibiting cyclin dependent kinases (CDKs). Examples of such compounds include butyrolactone I, flavopiridol, bohemin, olomoucine, roscovitine, purvanalol and indarubicine.
There is considerable support in the literature for the hypothesis that CDKs and their regulatory proteins play a significant role in the development of human tumours. Thus, in many tumours a temporal abnormal expression or activity of CDKs has been observed, together with a major deregulation of protein inhibitors (mutations, deletions). This results in the activation of CDKs and consequently in defective regulation of the Gl/S transition. Unlike normal cells, tumour cells do not arrest in Gl, and since they become independent of growth factors, they pass the Gl restriction point and enter the S phase very rapidly.
In contrast to the prior art, the present invention relates to compounds which are anti-proliferative, but which are believed to operate via a mechanism other than CDK inhibition.
The Gi/S transition of the mammalian cell cycle is tightly regulated by the retinoblastoma protein (pRb). Retinoblastoma gene mutations or deletions predispose individuals to familiar retinoblastoma and other types of cancers. The pRb protein is a docking protein, which in hypophosphorylated form has the capacity to bind and thus to inactivate S-phase transcription factors such as DP-1 and E2F. However, following phosphorylation by Gi/S cyclin-dependent kinases (CDKs) (CDK4/cyclin D1-D3, CDK6/cyclin D1-D3, CDK2/cyclin A), hyperphosphorylated pRb releases the transcription factors and S phase is initiated. Within the S phase, the pRb protein phosphorylation is maintained by the activity of CDK2/cyclin E complexes. Thus, hyperphosphorylation of the pRb protein plays a key role in the molecular pathology of cancer cells with altered CDK activity.
The present invention relates to the use of triterpenoid compounds derived from the natural products betulin and betulinic acid (BA) as shown in formula (A). The compounds of the present invention are referred to hereinafter as betulinines.
R = CH2OH: Betulin
R = COOH: Betulinic acid
(A)
With regard to their biological and therapeutic activity, the compounds disclosed herein are believed to be of specific benefit in the treatment of proliferative diseases such as cancers and leukaemias.
Several of the compounds suitable for use in the present invention are already known in the art, for example those disclosed in Ber. Dtsch. Chem. Ges. 55, 2332 (1922), Schulze H. et al; Acta Chem. Scand., B 29, 139 (1975), Suokas E. et al; Collect. Czech. Chem. Commun. 56, 2936 (1991), Sejbal J. et al; Collect. Czech. Chem. Commun. 64, 329 (1999), Klinotova E. et al; Indian. J. Chem., Sect. B 34, 624 (1995), Dinda B. et al. However, these disclosures do not include any indication as to possible biological activity of such compounds.
A first aspect of the present invention relates to the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, in therapy, in therapy,
wherein:
X1 is C=O, C=NORla, CHORla, CHOCORla, CHOC(O)ORu,
CHOC(O)ORla, CHOC(O)OR12, or CHOCOY-Hal
X4 is CH2, CH-Hal, C=O, CHORlb, CHOCORlb, CHOC(O)ORπ
XD is CH2, CH-Hal, C=O, CHOR l1bD, CHOCOR . l1b0, CHOC(O)OR , 11
R , 1-5 are H or lower alkyl
R7 is COORlc, COOR12, COHal, C(O)OC(O)Rlc, COOYOCORlc,
CH2ORlc, CH2OCORlc, CH2OC(O)ORπ
R9 is Rld, ORld, CH2-Hal, CH2ORld, CH2OC(O)ORn, =CHRld
R10is Rle, CH=NORle, CN, COORle, CORle, CH2-Hal, CH2ORle,
CH2OCORle, CH^C^OR1 CH2OSO2CH3, CH2OSO2C6H4CH3
R11 is an OH-substituted alkyl group, an ether group or a cyclic ether;
R12 is lower alkyl substituted by Hal
"b" is a double bond or a single bond and wherein Y = (CH2)n n = 0 to 5
Rla"lβare the same or different groups of R1 Hal is Cl, Br, I, F;
with the proviso that said compound is other than
3 β-3-hydroxyluρ-20(29)-en-28-oic acid;
3β-lup-20(29)-ene-3,28-diol;
3 β-lup-20(29)-ene3 ,28-diol diacetate;
3 β-3-(acetyloxy)luρ-20(29)-en-28-oic acid; 3β-3-(l-oxobutoxy)lup-20(29)-en-28-oic acid;
3β-luρ-20(29)-ene-3,28-diol 3-acetate;
3β-luρ-20(29)-ene-3,28-diol 28-acetate;
3β-3-hydroxylup-20(29)-en-28-oic acid methyl ester;
3β-3-(acetyloxy)lup-20(29)-en-28-oic acid methyl ester; 3 β-3- hydroxylup-20(29)-en-28-oic acid ethyl ester;
3 β-3- hydroxylup-20(29)-en-28-oic acid butyl ester;
3β-lupane-3,28-diol;
3 β-3- hydroxylupan-28-oic acid;
3 β-3- hydroxylupan-28-oic acid methyl ester; 3β-3-(acetyloxy)lupan-28-oic acid methyl ester;
3 β-3 -(acetyloxy)lupan-28-oic acid;
3β-lupane-3,28-diol diacetate;
3β-lupane-3,28-diol dibutanoate;
3β-3-(3-methyl-l-oxobutoxy)lupan-28-oic acid; 3β-3-(2,2-dimethyl-l~oxopropoxy)lupan-28-oic acid;
3β-3,28-dimethoxylup-20(29)-ene;
3 β-3,28-dimethoxylupane;
3 β-28-methoxylupan-3-ol;
3 β-3-methoxylup-20(29)-en-28-oic acid; 3β-3-methoxylup-20(29)-en-28-oic acid methyl ester; 3α-3-methoxylup-20(29)-en-28-oic acid; or 3 -3-methoxylup-20(29)-en-28-oic acid methyl ester.
In a preferred aspect, the invention relates to the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, for treating a patient suffering from leukaemia, cancer or other proliferative disorder.
A second aspect of the present invention relates to novel betulinines of structural formula la, or a pharmaceutically acceptable salt thereof,
wherein:
X1 is CHOC(O)ORn, CHOC(O)ORla, CHOC(O)OR12, or CHOCOY- Hal
X is CH2, CH-Hal, C=O, CHOR l1bD, CHOCOR , 11b0, CHOC(O)OR , 11
XD is CH2, CH-Hal, C=0, CHOR l1bD, CHOCOR , 11b0, CHOC(O)OR >n
R , 1-5 are H or lower alkyl R7 is COHal, C(O)OC(O)Rlc, COOYOCOR10, CH2OC(O)ORπ R9 is Rld, ORld, CH2-Hal, CH2ORld, CH2OC(O)ORu, =CHRld R10 is Rle, CH=NORle, CN, COORle, CORle, CH2-Hal, CH2ORle, CH2OCORle, CH2OC(O)ORπ, CH2OSO2CH3, CH2OSO2C6H4CH3 R11 is an OH-substituted alkyl group, an ether group or a cyclic ether; R12 is lower alkyl substitued by Hal "b" is a double bond or a single bond and wherein Y = (CH2)n n = 0 to 5
Rla"le are the same or different groups of R1 Hal is Cl, Br, I, F.
As used herein, the term lower alkyl means a linear or branched chain alkyl group containing from 1 to 6 carbon atoms, including methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl.
Description of the Preferred Embodiments Within the options provided for the groups X1, X4, X5 and R1"5, R7, R9"10 of formula I, the following options are preferred;
X1 is C=O, CHORla, CHOCORla, CHOCOY-Hal, CHOCOOR11; X4 is C=O, CH2; X5 is C=O, CH2, CHOCORlb;
R7 is COORlc, CH2ORlc, CH2OCORlc, COOYOCOR10, CH2OSO2C6H4CH3,
COHal, COOCOR10, CH2OCOORn;
R9 is Rld, =CHRld, CH2ORld, ORld;
R10 is Rle, CN, COORle, CH2ORle, CORle.
Preferably, R11 is a diol, triol or polyol.
Even more preferably, R1, R3"5 are methyl, R2 is H or methyl, and R1 is defined as below for the relevant group Rla"e; when X1 is:
-CHORla, Rla is H; -CHCOY-Hal, Y is CH2 and Hal is Cl; -CHOCORla, Rla is methyl;
-CHOCOOR11, R11 is glyceryl, 2,2-dimethyl-l,3-dioxolan-4-yl or pentaerythrityl; when X5 is CHOCORlb, Rlb is methyl; when R7 is:
-COORlc, Rlc is H or methyl; -COOYOCOR10, Y is CH2 and Rlcis H; -CH2ORlc, Rlc is H;
-CH2OCORIc, Rlc is methyl;
-COOCOR10, Rlc is methyl or butyl;
-COHal, Hal is Cl;
-CH2OCOORn, R11 is glyceryl, 2,2-dimethyl-l,3-dioxolan-4-yl or pentaerythrityl; when R9 is =CHRld, CH2ORld or ORld, Rld is H, and when R9 is Rld, Rld is methyl; when R10 is Rle, Rle is methyl, and when R10 is COORle, CORle or CH2ORle, Rle is H.
In a more preferred embodiment of the first aspect of the invention, the compounds of use are selected from those shown in Table 1 below.
Table 1
where: A is CHOC(O)OCH2CHOHCH2OH
B is CHOC(O)OCH2-2,2-dimethyl-l,3-dioxolan-4-yl Ts is OSO2C6H4CH3.
In a particularly preferred embodiment, said compound is selected from the following:
In respect of the second aspect of the invention, the preferred embodiments regarding the compounds are identical to those given above for the first aspect with application of the proviso of formula la.
The most preferred compounds of the second aspect are those in Table la. Table la
where: A is CHOC(O)OCH2CHOHCH2OH
B is CHOC(O)OCH2-2,2-dimethyl-l,3-dioxolan-4-yl Ts is OSO2C6H4CH3.
In respect of the invention as a whole, it is preferable that the proliferative disorder is cancer or leukaemia. In one embodiment, the cancer or leukaemia is p53, hormone and multidrug resistance independent. In another embodiment, the cancer or leukaemia is independent of Rb status.
More specifically, the present invention relates to a method of treating patients suffering from cancer by administering therapeutically effective amounts of a compound of formula I or pharmaceutically acceptable salts or esters thereof.
Without wishing to be bound by theory, preliminary studies suggest that rather than influencing the activity of cyclin dependent kinases, the compounds of the present invention appear to operate via an alternative mechanism. In particular, it is believed that the betulinines of the present invention may inhibit cell proliferation and induce cancer cell death in a manner which involves mainly post-translational modifications, namely the phosphorylation, of a key regulatory protein involved in cellular proliferation. More specifically, it is believed that the betulinines of the invention effect a change in the phosphorylation state of the Rb protein. Such a mechanism may be advantageous as it is thought that the compounds of the present invention may be capable of inhibiting cell proliferation in proliferating tumour tissue, but not in healthy tissue.
Thus, in a further embodiment the present invention relates to a method of treating a cancerous or leukaemic proliferative disease through effecting a change in the pRb protein phosphorylation state by the administration of a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salts or esters thereof.
The compounds of the present invention are also capable of inducing apoptosis (programmed cell death) in proliferative cells. Thus, in an additional embodiment, the present invention relates to a method of inducing cell death in proliferative cells comprising administering a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salts or esters thereof.
A further aspect of the present invention relates to use of betulinines of formula I as research chemicals and as compounds for clinical and/or laboratory diagnostics. More particularly, the invention relates to the use of betulinines as research chemicals for studying the phosphorylation/de-phosphorylation processes of cellular substrates, cellular proliferation, purification of target molecules, and/or cell cycle studies. The present invention therefore further relates to the use of a compound of formula I in the manufacture of a medicament for use in the treatment of a proliferative disease.
As used herein the phrase "manufacture of a medicament" includes the use of a compound of formula I directly as the medicament in addition to its use in a screening programme for further anti-proliferative agents or in any stage of the manufacture of such a medicament.
Such a screening programme may for example include an assay for determining the phosphorylation state of cellular substrates and determining whether a candidate substance is capable of mimicking the activity of a betulinine of formula I.
Thus, in a further embodiment, the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt, crystal form, complex, hydrate, or hydrolysable ester thereof, in an assay for determining the phosphorylation state of cellular substrates, and optionally in the identification of candidate compounds that act in a similar manner.
Preferably, the cellular substrate, the phosphorylation state of which is being assayed is Rb protein.
Such assays may be carried out by incubating a betulinin either alone or together with a candidate substance with a relevant cell line and assessing the phosphorylation profile the Rb protein over a period of time. If a candidate substance is present it's effect on the activity of the control betulinin will be evident by running the corresponding controls (betulinin alone and candidate alone). Further information on such assays including appropriate cell lines,reagents and Rb antibodies is given below. Rb phosphorylation assay;
Since Rb protein contains multiple phosphorylation sites for CDKs, its phosphorylated form has molecular weight about 110 kDa, while the molecular weight of hypophosphorylated protein is only 105 kDa. This small difference in molecular weight is enough to separate both forms by conventional SDS-PAGE electrophoresis.
CEM cells may are cultured in Dulbeco's modified essential medium with 4.5 g dextrose/1, 10% of foetal calf serum, 2 mM glutamine, 100 U/ml penicillin and 100 μg/ml streptomycin with/without below indicated concentrations of betulinin. At selected time points, cells are harvested, washed in ice cold Hank's balanced salt solution and solubilized on ice using the SDS-PAGE sample buffer containing protease and phosphatase inhibitors (10 μg/ml of leupeptin, 10 μg/ml of aprotinin, 10 μg/ml of soybean trypsin inhibitor, 100 μmol of benzamide, 1 mM of sodium vanadate, 1 mM of NaF, 1 mM of phenylphosphate) and boiled immediately.
Total cellular proteins (100 μg/well) are separated on SDS-PAGE electrophoresis, blotted on polyvinyldifluoride membranes and total Rb protein, including proteolytic fragment(s) detected using a pRb monoclonal antibody (Oncogene, Germany, Rb(Ab-5), Cat# OP66 Rev 02-Sept-96 EB, Clone LM95.1) and visualized by chemiluminiscence (ECL- estern Blotting System, Amersham). Details of the Western blot technique are described in Ausubel, F.M., Brent, R., Kingston, R.E., Moore, D.D., Seidman, J.G., Smith, J.A., Struhl, K (Eds): Short Protocols in Molecular Biology, 2nd edition, John Wiley & Sons, New York, Chichester, Brisbane, Toronto, Singapore, 1992, page 10-33 - 10-35.
The compounds of the present invention can be present as salts or esters, in particular pharmaceutically acceptable salts or esters. Pharmaceutically acceptable salts of the product of the invention include suitable acid addition or base salts thereof. A review of suitable pharmaceutical salts may be found in Berge et al, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for. example with strong inorganic acids such as mineral acids, e.g. sulphuric acid, phosphoric acid or hydrohalic acids; with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Ci-C )-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p- toluene sulfonic acid.
Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esterified. Organic acids include carboxylic acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acid, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as ( -G -alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid. Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide. Alcohols include alkanealcohols of 1-12 carbon atoms which may be unsubstituted or substituted, e.g. by a halogen). In all aspects of the present invention previously discussed, the invention includes, where appropriate all enantiomers and tautomers of compounds of formula I or la. The man skilled in the art will recognise compounds that possess optical properties (one or more chiral carbon atoms) or tautomeric characteristics. The corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art.
The invention furthermore relates to the compounds of, or of use, in the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms by slightly varying the method of purification and or isolation from the solvents used in the synthetic preparation of such compounds.
The invention further includes the compounds of, or of use, in the present invention in prodrug form. Such prodrugs are generally compounds of formula I or la wherein one or more appropriate groups have been modified such that the modification is reversed upon administration to a human or mammalian subject. Such reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo. Examples of such modifications include esters (for example, any of those described above), wherein the reversion may be carried out be an esterase etc. Other such systems will be well known to those skilled in the art.
The present invention also encompasses pharmaceutical compositions comprising the compounds of the invention. In this regard, and in particular for human therapy, even though the compounds of the present invention (including their pharmaceutically acceptable salts, esters and pharmaceutically acceptable solvates) can be administered alone, they will generally be administered in admixture with a pharmaceutical carrier, excipient or diluent selected with regard to the intended route of administration and standard pharmaceutical practice.
Thus, the present invention also relates to pharmaceutical compositions comprising betulinines or pharmaceutically acceptable salts or esters thereof, together with at least one pharmaceutically acceptable excipient, diluent or carrier.
By way of example, in the pharmaceutical compositions of the present invention, the compounds of the invention may be admixed with any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), and/or solubilising agent(s). Examples of such suitable excipients for the various different forms of pharmaceutical compositions described herein may be found in the "Handbook of Pharmaceutical Excipients, 2nd Edition, (1994), Edited by A Wade and PJ Weller.
The pharmaceutical compositions of the present invention may be adapted for oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual routes of administration.
For oral administration, particular use is made of compressed tablets, pills, tablets, gellules, drops, and capsules. Preferably, these compositions contain from 1 to 250 mg and more preferably from 10-100 mg, of active ingredient per dose.
Other forms of administration comprise solutions or emulsions which may be injected intravenously, intraarterially, intrathecally, subcutaneously, intradermally, intraperitoneally or intramuscularly, and which are prepared from sterile or sterilisable solutions. The pharmaceutical compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
An alternative means of transdermal administration is by use of a skin patch. For example, the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin. The active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
Injectable forms may contain between 10 - 1000 mg, preferably between 10 - 250 mg, of active ingredient per dose.
Compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
A person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation. Typically, a physician will determine the actual dosage which will be most suitable for an individual patient and it will vary with the age, weight and response of the particular patient. The dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
In an exemplary embodiment, one or more doses of 10 to 150 mg/day will be administered to the patient for the treatment of malignancy. The invention further relates to methods of chemical synthesis of the above described compounds.
In one embodiment, the invention relates to a process for preparing a compound of formula I, as defined above, wherein X4 is C=O and X5 is C=O or CH2,
comprising
(i) oxidising a compound of formula lb to form a compound of formula Ic; and (ii) where Rlc is H, esterifying with an appropriate group bearing the desired Rlc substituent; and optionally (iii) oxidising the product of step (i) or (ii) to form a compound of formula Id.
In a preferred embodiment, the compound of formula lb is oxidised to Ic by treating with ruthenium tetroxide.
In a preferred aspect, the compound of formula Ic is oxidised to Id by treating with selenium dioxide.
A further embodiment of the invention relates to a process for preparing a compound of formula I as defined above, wherein X4 and X5 are CH ,
comprising oxidising a compound of formula le to form a compound of formula
If.
In a preferred aspect, the compound of formula le is oxidised to If by treating with selenium dioxide.
The preparation of the compounds of the present invention will be discussed in greater detail below, with specific reference to the preferred embodiments. The man skilled in the relevant art would be able to prepare other compounds of the invention by selection of the appropriate reagents.
The following scheme illustrates the synthesis of compounds of formula I where X1 is CHOAc, X4 is C=O, X5 is CH2 or C=O, R1"5'9'10 are methyl, and R7 is CH2OAc, COOMe or COOCH2OCOC(CH3)3.
C(CH3)3 Conditions: a, hydrolysis (e.g. with potassium hydroxide); b, oxidation (e.g. with ruthenium tetroxide); c, esterification (e.g. with diazomethane); d, esterification with POM-Cl in the presence of base (e.g. DBU); e, esterification with AcM-Br in the presence of base (e.g. DBU); f, oxidation (e.g. with selenium dioxide).
The scheme below illustrates the synthesis of compounds of formula I where X1 is CHOAc, X4'5 are CH2, R1-5 are methyl, R7 is COOH, R9 is CH2, and R10 is COH.
Conditions: a, oxidation (e.g. with selenium dioxide).
The scheme below illustrates the synthesis of compounds 11.34, 11.35, 11.39 - 11.48, 11.50 and 11.51 of formula I where R1'5 are methyl.
ig In
_
X = C=θ, X° = CH2 = C=θ, X = CH2 X 4* = C=θ, Xs = CH2 R9 = CH3, R10 = CH3 = CH3, R = CH3 = CH3, R = CH3 R7 = COOCH3 = COOCH3 = C00CH3 b is double bond is double bond is double bond
X4 = C=θ, Xs = CH2 X4 = C=θ, Xs = CH2 X4 = C=θ, X5 = CH2 R9 = CH3, R10 = CH3 : CH3, R — CH3 R7 = COOH • COOH =>7 = COOH b is double bond b is double bond b is double bond
X4 = C=0, X° = C=θ = c=o, xb = c=o = C=0, X5 = C=θ
?9 = R9 = CH3, R10 = CH3 CH3f R = CH3 : CH , R - CH3 R7 = COOCH3 = COOCH3 = COOCH3 b is double bond b is double bond b is double bond
X — CH , X — CH X" = X — CH21 X - CH R9 = =CH2, R10 = CH R9 = = =CH2, R CH, = =CH2, R10 = CH3 R7 = COOH R = = COOH = COOH b is single bond b is single bond is single bond
X =2, X = CH2 = CH2, X5 = CH2 *« = CH , X -= CH R9 = =CH2, R10 = CH3 — CH2, R = CH3 = =2l R = CH R7 = COOCH3 R' = COOCH3 = COOCH3 b is single bond b is single bond b is single bond
X = CH2, X — CH2 X4 = f CH , X — CH2 R9 = =CH2, R10 = CH3 — — πi R — CH3 59 = =CH2, R10 = CH3 R7 = COOCH2OCOCH3 = COOCH2OCOCH3 = COOCH2OCOCH3 b is single bond » single bond b is single bond or
OH
Conditions: a, esterification with chloroformate of solketal in presence of base (e.g. pyridine); b, deprotection of acetonide (e.g. with hydrochloric acid). The scheme below illustrates the synthesis of compounds 11.54 - 11.58 of formula I where R*-R5 are methyl, X5 is CH2, X1 is CHOAc or C=O, X4 is CH2 or C=O, R9 is =CH2 or CH3, R10 is CH3 or COH and R7 is COOH or COF.
X1 = CHOAc, X4 = CH2 X1 = CHOAc, X" = CH2 R9 = =CH2, RW = CH3 R9 = =CH2, R10 = CH3 b is single bond b is single bond or or r
X1 = CHOAc, X4 = CH2 X1 = CHOAc, X" = CH2 R9 = CH3, R1° = CH3 R9 = CH3, R 0 = CH3 b is single bond b is single bond or
X' = C=0, X" = CH2 X1 = C=0, X4 = CH2 R9 = =CH2, R« = CH3 R9 = =CH2, Ri° = CH3 b is single bond b is single bond or or
X1 = CHOAc, X4 = CH2 X1 = CHOAc, X4 = CH2 R9 = =CH2, R1° = COH R9 = =CH2, R10 = COH b is single bond b is single bond or
X1 = CHOAc, X4 = C=0 X1 = CHOAc, X4 = C=0 R9, R10 = CH3 R9, R 0 = CH3 b is double bond b is double bond
Conditions: a, reaction with diethylaminosulphur trifluoride.
This invention is further illustrated by the following examples, which should not be construed as further limiting. EXAMPLES General
The chemical shift values (δ-scale, ppm) and coupling constants (J, Hz) in the 1H and 13C NMR spectra were obtained using a Varian UNITY-INOVA 400 FT spectrometer (1H at 400 MHz and 13C at 100.6 MHz) in deuterochloroform with tetramethylsilane (for 1H NMR data δ = 0 ppm) as an internal standard. For the 13C NMR data δ(CDCl3)=77.00 ppm. The value for a multiple!, either defined (doublet (d), triplet (t), quartet (q), septet (sept.) or not (m) at the approximate mid point is given unless a range is quoted (s=singlet, b=broad)).
Electron impact mass spectra (EIMS) were measured on an INCOS 50 instrument. Ionising electron energy 75 eV, ion source temperature 150 °C.
EIMS was used to determine molecular weights, M1" corresponding to the molecular ion.
Ether is diethylether. THF and dioxane were dried over sodium. Acetic acid was purified before use by chromium trioxide treatment and distillation. Reactions were run at room temperature unless otherwise stated. The reaction progress was monitored by thin layer chromatography (TLC) on silicagel 60 G (Merck, detection by spraying with 10 % sulphuric acid and heating). The work-up procedure involves dilution with specified solvent (otherwise the organic reaction solvent), extraction with water and then brine or sodium hydrogencarbonate, drying over anhydrous magnesium sulphate, and evaporation under vacuum to give a residue.
EXAMPLE 1
Lup-20(29 -ene-3 β.28-diyl diacetate
Crude betuline (500 g) was dissolved in a mixture of 250 ml pyridine and 250 ml acetic anhydride.. The mixture was then refluxed for half an hour. After cooling, the resulting crystals were filtered off and washed with acetic acid, ethanol and water. A solution of crude lup-20(29)-ene-3β,28-diyl diacetate (400 g) in chloroform was filtered through a column of alumina, and the column was washed with chloroform. The filtrate was then evaporated under reduced pressure. The residue was crystallized from chloroform/methanol to obtain 250 g of the title compound which according to TLC contained traces of lupeol acetate. After recrystallization from chloroform/methanol, the yield of pure compound was 239 g, mp. 222 - 223 °C, [α]D+22° (c 0.4; CHC13). [Schulze H., Pieroh K.: Ber. Dtsch. Chem. Ges. 55, 2332 (1922)].
The 1H NMR spectrum of the title compound is as follows:
0.84 s, 0.84 s, 0.85 s, 0.97 s, 1.03 s, 1.68, 6 x 3H (6 x CH3); 2.04 s, 3 H, 2.07 s, 3 H (2x OAc); 2.44 ddd, 1 H (J = 11.4, J" = 10.9, Y" = 0.7, H-19); 3.85 d, 1 H (J = 11.1, H-28a); 4.25 dd, 1 H, (J' = 11.1, J" - 1.4, H-28b); 4.47 m, 1 H (H- 3α); 4.59 m, 1 H (Σ J = 3.4, H-29E); 4.69 m, 1 H (Σ J = 2.1, H-29Z).
EXAMPLE 2
Lup- 18-ene-3 β ,28-diyl diacetate
A solution of hydrogen bromide in acetic acid (38 %, 1.4 1) was added to a solution of lup-20(29)-ene-3β,28-diyl diacetate (100 g, 190 mmol) in a mixture of benzene, acetic acid and acetic anhydride (1 1 : 0.5 1 : 50 ml). The reaction mixture was refiuxed until the reaction was completed (TLC was developed in hexane/ether mixture). After cooling, the reaction mixture was poured into ice cold water (3:1) and extracted with benzene (3 x 0.5 1). The combined organic phases were washed with NaHCO3 aqueous solution, NaHSO3 solution and water and dried over magnesium sulphate. Usual working up procedure gave 90 g of dark brown residue. The dry powder was extracted in a Soxhlet extractor with acetone until it turned white. After drying in the air, the product was crystallized from butanone. The yield of the title compound was 74 g (74 %), mp. 215-216 °C, [α]D +15° (c 0.45; CHC13). [Suokas E., Hase T.: Acta Chem. Scand., B 29, 139 (1975)].
The 1H NMR spectrum of the title compound is as follows: 0.84 s, 0.85 s, 0.89 s, 0.90 s, 0.91 d, 3 H (J = 6.8), 0.99 d, 3 H (J = 6.8), 1.06 s, 7 x 3H (7 x CH3); 2.04 s, 3 H, 2.05 s, 3 H (2x OAc); 2.25 m, 2 H (Σ J ~ 15); 2.43 m, 1 H (Σ J ~ 15); 3.14 sept, 1 H (J = 7, H-20); 3.98 d, 1 H (J = 10.8, H-28a); 4.03 d, 1 H (J = 10.8, H-28b); 4.49 m, 1 H (H-3α).
EXAMPLE 3
21 -oxo-lup- 18-ene-3 β,28-diyl diacetate
Lup-18-ene-3β,28-diyl diacetate (50 g; 95 mmol), sodium dichromate (22.5 g;
75.5 mmol) and sodium acetate (5 g) were dissolved in a mixture of benzene and acetic acid (0.7 1, 0.3 1). The reaction mixture was allowed to stand until the reaction was completed (TLC was developed in hexane/ether). After dilution with an excess of water, the mixture was extracted with benzene (3 x 300 ml).
After usual working up procedure the title compound was obtained (45 g, 87 %) as a pale-yellow crystalline foam which was used in the next step without further purification (see Example 4). Pure title compound has m.p. 205 - 206 °C, [α]D -35° (c 0.49; CHC13). Another way to the title compound is described in Sejbal J., Klinot J., Budes nsky M., Protiva J.: Collect. Czech. Chem.
Commun. 56, 2936 (1991).
The 1H NMR spectrum of the title compound is as follows: 0.85 s, 0.86 s, 0.93 s, 0.94 s, 1.16 s, 1.17 d (J = 7.1), 1.21 d (J = 7.1), 7 x 3 H (7x CH3); 2.00 s, 3 H, 2.05 s, 3 H (2x OAc); 2.39 d, 1 H (J= 18.5, H-22); 2.87 dd, 1 H (J '= 11.9, J" = 4.1, H-13β ); 3.18 sept, 1 H (J = 6.6, H-20); 4.06 d, 1 H (J = 10.9, H-28a); 4.34 d, 1 H (J = 10.9, H-28b); 4.49 m, 1 H (J ~ 7, H-3α). The following compounds were prepared by the above-mentioned procedure: (pivaloyloxy)methyl 3 β,28-diacetoxy-l 8-oxo- 19,20,21 ,29,30-pentanorlupan- 22-oate acetoxymethyl 3 β,28-diacetoxy- 18-oxo- 19,20,21 ,29,30-pentanorlupan- 22-oate
EXAMPLE 4
21 ,22-dioxolup- 18-ene-3 β ,28-diyl diacetate
A solution of crude 21-oxolup-18-ene-3β,28-diyl diacetate (40g; 74 mmol; containing about 85 % of 21-oxo-lup-18-ene-3β,28-diyl diacetate) and selenium dioxide (160 g; 1.44 mol) in a mixture of dioxane (0.8 1) and acetic acid (0.4 1) was refluxed until the reaction was completed (TLC was developed in benzene/ether).
After cooling, the precipitated selenium was removed by filtration and the filtrate was slowly poured into a vigorously stirred excess of water. The red- orange precipitate was filtered off under reduced pressure, carefully washed with water and dried in the air. Dry crude 21,22-dioxo-lup-18-ene-3β,28-diyl diacetate was dissolved in chloroform and the solution was filtered through a column of alumina, the column was then washed with chloroform, and the filtrate was evaporated under reduced pressure. The residue was crystallized from methyl acetate to give 28.9 g (82 %) of the title compound as pale-orange needles, mp. 267-270 °C, [α]D -127° (c 0.32; CHC13). Another way to the title compound is described in Klinotova E., Cermakova J., Rejzek M., Krecek V., Sejbal J., Olsovslcy P., Klinot J.: Collect. Czech. Chem. Commun. 64, 329 (1999).
The 1H NMR spectrum of the title compound is as follows:
0.85 s, 0.86 s, 0.94 s, 0.97 s, 1.18 s, 1.24 d (J = 7.2), 1.26 d (J = 7.2), 7 x 3 H
(7x CH3); 1.93 s, 3 H, 2.06 s, 3 H (2x OAc); 3.12 dd, 1 H (J '= 12.5, J" = 3.8, H-13β ); 3.36 sept, 1 H (J = 7.0, H-20); 4.02 d, 1 H (J = 11.1, H-28a); 4.49 dd, 1 H (J' = 10.2, J" = 6.0, H-3α); 4.84 d, 1 H (J = 11.1, H-28b).
The following compounds were prepared by the above-mentioned procedure: methyl 3 β-acetoxy-21 ,22-dioxolup- 18-en-28-oate acetoxymethyl 3 β -acetoxy-21 ,22-dioxolup- 18-en-28-oate
(pivaloyloxy)methyl 3 β-acetoxy-21 ,22-dioxolup- 18-en-28-oate acetoxymethyl-3 β,28-diacetoxy- 18-oxo- 19,20,21 ,29,30-pentanorlup- 12-en-22- oate (pivaloyloxy)methyl 3 β,28-diacetoxy- 18-oxo- 19,20,21 ,29,30-pentanorlup- 12- en-22-oate
3β-hydroxy-30-oxolup-20(29)-en-28-oic acid [Dinda B., Hajra A. K., Das S. K,
Chel G., Chakraborty R., Ranu B. C: Indian. J. Chem., Sect. B 34, 624 (1995)]. acetoxymethyl 3 β-hydroxy-30-oxolup-20(29)-en-28-oate (pivaloyloxy)methyl 3β-hydroxy-30-oxolup-20(29)-en-28-oate
EXAMPLE 5
28-hydroxy-21 -oxolup- 18-en-3 β-yl acetate
A solution of 21-oxolup-18-ene-3β,28-diyl diacetate (20 g; 37 mmol) and potassium hydroxide (2.5 g; 45 mmol) in a mixture of benzene (200 ml) and ethanol (200 ml), was vigorously stirred until the reaction was completed (TLC was developed with the mixture hexane/ether).
The mixture was then poured into dilute (1 :4) hydrochloric acid and extracted with benzene (3 x 200 ml). The organic layer was processed by the usual working up procedure to yield 17.5 g (95%) of a snow-white crystalline residue. Pure title compound has m.p. 292 - 294 °C, [α]D -69° (c 0.34; CHC13). [Klinotova E., Cermakova J., Rejzek M., Krecek V., Sejbal J., Olsovsky P., Klinot J.: Collect. Czech. Chem. Commun. 64, 329 (1999)]. The 1H NMR spectrum of the title compound is as follows: 0.85 s, 0.86 s, 0.93 s, 0.95 s, 1.13 s, 1.19 d (J = 6.9), 1.21 d (J = 6.9), 7 x 3 H (7x CH3); 1.92 d, 1 H (J = 18.6, H-22a); 2.05 s, 3 H (OAc); 2.44 d, 1 H (J= 18.6, H-22b); 2.78 dd, 1 H (J '= 12.5, J" = 3.4, H-13β ); 3.19 sept, 1 H (J = 6.9, H-20); 3.67 d, 1 H (J = 10.7, H-28a); 3.72 d, 1 H (J = 10.7, H-28b); 4.49 dd, 1 H (J'= 11.0, V = 5.5, H-3α).
EXAMPLE 6
17β-methoxycarbonyl-28-norlup-20(29)-en-3 β-yl r(2.2-dimethyl-l .3-dioxolan- 4-yl methyl] carbonate
A solution of methyl betulinate (0.5 g; 1.1 mmol) in a mixture of dichloromethane (8.0 ml), pyridine (0.4 ml) and chloroformate of solketal (0.8 ml; 4.1 mmol) was vigorously stirred for twelve hours at room temperature. After the reaction was complete the mixture was slowly poured into 100 ml of 5 % hydrochloric acid and ice, twice extracted with chloroform. The combined organic layers were worked up in the usual manner and chromatographed on silica gel (10 % ethyl acetate in hexane). After crystallization from methanol, the yield of pure compound was 0.45 g (67 %), mp. 106 - 108 °C, [α]D +17° (c 0.56; CHC13).
The 13C NMR spectrum of the title compound is as follows: 176.7, 155.1, 109.9, 109.6, 150.5, 85.8, 73.5, 73.4, 67.5, 66.4, 56.6, 55.4, 51.3, 50.4, 49.4, 47.0, 42.4, 40.7, 38.3, 38.2, 38.0, 37.1, 36.9, 34.2, 32.1, 30.6, 29.7, 27.8, 26.7, 25.5, 25.4, 22.6, 20.9, 19.3, 18.1,16.4, 16.1, 15.9, 14.7.
The following compounds were prepared by the above-mentioned procedure: 17β-mefhoxycarbonyl-21 -oxo-28-norlup- 18-en-3 β-yl [(2,2-dimethyl-l ,3-dioxo- lan-4-yl)methyl] carbonate
17β-methoxycarbonyl-21 ,22-dioxo-28-norluρ- 18-en-3 β-yl [(2,2-dimethyl- 1 ,3 - dioxolan-4-yl)methyl] carbonate 17β-carboxy-21-oxo-28-norlup-18-en-3β-yl[(2,2-dimethyl-l,3-dioxolan-4- yl)methyl] carbonate
17β-carboxy-28-norlup-20(29)-en-3β-yl [(2,2-dimethyl- 1, 3 -dioxolan-4-yl)met- hyl] carbonate 17β-(acetoxymethylen)oxycarbonyl-28-norlup-20(29)-en-3 β-yl [(2,2-dimethyl- 1 ,3-dioxolan-4-yl)methyl] carbonate
17β-methylene-28-norlup-20(29)-en-3 β-yl bis [(2,2-dimethyl- 1 ,3-dioxolan-4- yl)methyl] dicarbonate
EXAMPLE 7
17β-methoxycarbonyl-28-norlun-20('29 -en-3β-yl(2.3-dihvdroxypropyD carbonate
A solution of HCI (10 %; 6.0 ml) was added to a - solution of l7β- methoxycarbonyl-28-norlup-20(29)-en-3β-yl[(2,2-dimethyl-l,3-dioxolan-4- yl)methyl] carbonate (0.4 g; 0.6 mmol) in THF (8.0 ml) and the clear reaction mixture was stirred for five hours at 30 °C. After the reaction was complete, the mixture was diluted with cold water (50 ml) and twice extracted with ethylacetate. The combined organic layers were worked up in the usual manner and chromatographed on silica gel (toluene/ethyl acetate, 5:1). The yield of white lyophilized (from acetic acid) powder was 0.28 g (76 %), m.p. 89 - 91 °C, [α]D +14° (c 0.58; CHC13).
The 13C NMR spectrum of the title compound is as follows: 176.7, 155.6, 150.5, 109.6, 86.2, 70.1, 68.4, 63.2, 56.5, 55.4, 51.3, 50.4, 49.4, 47.0, 42.4, 40.7, 38.3, 38.2, 38.1, 37.1, 37.0, 34.2, 32.1, 30.6, 29.6, 27.9, 25.4, 23.6, 20.9, 19.3, 18.1, 16.4, 16.1, 15.9, 14.7.
The following compounds were prepared by the above-mentioned procedure: 17β-methoxycarbonyl-21-oxo-28-norlup-18-en-3β-yl(2,3-dihydroxypropyl) carbonate 17β-methoxycarbonyl-21 ,22-dioxo-28-norlup- 18-en-3 β-yl(2,3-dihydroxypro- pyl) carbonate
17β-carboxy-21 -oxo-28-norlup- 18-en-3 β-yl (2,3 -dihydroxypropyl) carbonate 17β-carboxy-28-norlup-20(29)-en-3β-yl (2,3-dihydroxypropyl) carbonate 17β-(acetoxymethylen)oxycarbonyl-28-norlup-20(29)-en-3β-yl (2,3 -dihydroxypropyl) carbonate
17β-methylene-28-norlup-20(29)-en-3β-yl bis(2,3 -dihydroxypropyl) di- carbonate
EXAMPLE 8
3 β-acetoxy-luρ-20(29V en-28-oyl fluoride
Diethylaminosulphur trifluoride (0.2 ml; 1.3 mmol) was added dropwise to a solution of acetylbetulinic acid (0.2 g; 0.4 mmol) in dry chloroform (2 ml) and the reaction mixture was stirred for one hour at room temperature. After the reaction was complete, the mixture was slowly poured into cold water (50 ml) and twice extracted with chloroform. The combined organic fractions were worked up in the usual manner and chromatographed on silica gel (3 % ethyl acetate in hexane). The residue was crystallized from isopropyl alcohol to obtain 0.13 g (65 %) of the title compound as small white crystals, m.p. 211 - 214 °C (decomp.), [α]D +38° (c 0.44; CHC13).
The 13C NMR spectrum of the title compound is as follows: 171.0 s, 165.2 d (J = 375), 149.3 s, 110.4 s, 80.9 s, 57.0 d (J = 39), 55.4 s, 50.4 s, 49.1 s, 46.7 s, 42.4 s, 40.7 s, 38.5 s, 38.4 s, 37.8 s, 37.1 s, 35.6 s, 34.2 s, 30.9 s, 30.0 s, 29.7 s, 27.9 s, 25.3 s, 23.7 s, 21.3 s, 20.8 s, 19.3 s, 18.1 s, 16.5 s, 16.2 s, 15.9 s, 14.7 s.
The following compounds were prepared by the above-mentioned procedure: 3β-acetoxy-21-oxolup-18-en-28-oyl fluoride 3β-acetoxylup -28-oyl fluoride 3-oxolup-20(29)-en-28-oyl fluoride 3 β-acetoxy-30-oxoluρ-20(29)-en-28-oyl fluoride
EXAMPLE: 9 BIOLOGICAL ACTIVITY OF BETULININES
In vitro cytotoxic activity of betulinines on tumor cell lines One of the parameters used as the basis for colorimetric assays is the metabolic activity of viable cells. For example, a microtiter assay which uses the tetrazolium salt MTT is now widely used to quantitate cell proliferation and cytotoxicity [Hajdύch M, Mihal V, Minarik J, Faber E, Safarova M, Weigl E, Antalek P.: Cytotechnology, 1996, 19, 243-245]. For instance, this assay is used in drug screening programs and in chemosensitivity testing. Because tetrazolium salts are cleaved only by metabolically active cells, these assays exclusively detect viable cells. In the case of the MTT assay, yellow soluble tetrazolium salt is reduced to a coloured water-insoluble formazan salt. After it is solubilized, the formazan formed can easily and rapidly be quantified in a conventional ELISA plate reader at 570 nm (maximum absorbancy). The quantity of reduced formazan corresponds to the number of vital cells in the culture.
Human T-lymphoblastic leukaemia cell line CEM was used for routine screening of these compounds. To prove a common mechanism of action, selected compounds which showed activity in a screening assay were tested in a panel of cell lines (Table 2). These lines were from different species and of different histogenetic origin and they possess various alterations in cell cycle regulatory proteins and hormone dependence status (Table 2). The cells were maintained in Nunc/Corning 80 cm2 plastic tissue culture flasks and cultured in cell culture medium (DMEM with 5 g/1 glucose, 2mM glutamine, 100 U/ml penicillin, 100 μg/ml streptomycin, 10% foetal calf serum and sodium bicarbonate). Individual compounds were dissolved in 10% dimethylsulfoxide/saline, pH 8.0.
The cell suspensions that were prepared and diluted according to the particular cell type and the expected target cell density (2.500-30.000 cells per well based on cell growth characteristics) were added by pipette (80μl) into 96/well microtiter plates. Inoculates were allowed a pre-incubation period of 24 hours at 37°C and 5% CO2 for stabilisation. Four-fold dilutions of the intended test concentration were added at time zero in 20μl aliquots to the microtiter plate wells. Usually, test compounds were evaluated at six 4-fold dilutions. In routine testing, the highest well concentration was 250 μM, but it may differ, depending on the agent. All drug concentrations were examined in duplicate. Incubations of cells with the test compounds lasted for 72 hours at 37°C, in 5% CO2 atmosphere and 100% humidity. At the end of the incubation period, the cells were assayed by using the MTT assay. Ten microliters of the MTT stock solution were pipetted into each well and incubated further for 1-4 hours. After this incubation period, formazan was solubilized by the addition of lOOμl/well of 10% SDS in water (pH=5.5) followed by further incubation at 37°C overnight. The optical density (OD) was measured at 540nm with the Labsystem iEMS Reader MF(UK). The tumour cell survival (TCS) was calculated using the following equitation: exposed well / mean ODrøntroi wells) 100%. The TCS50 value, the drug concentration lethal to 50% of the tumour cells, was calculated from the obtained dose response curves.
To evaluate the . anti-cancer activity of betulinines, their cytotoxic activity against CEM cell line was examined using the screening assay. Potent compounds were further tested against a panel of cell lines of different histogenetic and species origin (Table 2). Table 2 Cytotoxic activity of selected betulinines against a panel of different (non)malignant cell lines.
In contrast to betulinic acid, which is reported to be an agent selective for neuroectodermal derived tumours, there was no significant difference in sensitivity of betulinines to tumours of different histogenetic origin.
The compounds are effective in submicromolar or low micromolar concentrations. However, the non-malignant cells, e.g. NIH3T3 fibroblasts and normal human lymphocytes, tolerated substantially higher doses of betulinines than the tumour cells suggesting a favourable therapeutic index.
Notably, the effectiveness of betulinines was found to be identical in cell lines bearing various mutations or deletions in cell cycle associated proteins (Table 2). This indicates that these substances should be equally effective in tumours with various alterations of tumour suppresser genes, namely p53, Rb, etc.
Furthermore, betulinines were shown to be equally effective in drug resistant cell lines as on their maternal counterparts, thereby suggesting that classical mechanisms of multidrug resistance apparently do not apply to these compounds. This particular characteristic should be of significant therapeutic benefit to chemotherapy resistant cancer patients.
Finally, the cytotoxic activity of betulinines is independent of the hormonal status of cancer cells, so the compounds should be equally effective in treatment of hormone dependent and independent cancers.
Those skilled in the art will recognise, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the claims.

Claims

Use of a compound of formula I, or a pharmaceutically acceptable salt thereof, in therapy,
wherein:
X1 is C=O, C=NORla, CHORla, CHOCORla, CHOC(O)ORn,
CHOC(O)ORla, CHOC(O)OR12, or CHOCOY-Hal
X4 is CH2, CH-Hal, C=O, CHORlb, CHOCORlb, CHOC(O)ORn
X5 is CH2, CH-Hal, C=O, CHORlb, CHOCORlb, CHOC(O)ORπ
R1"5 are H or lower alkyl
R7 is COORIC, COOR12, COHal, C(O)OC(O)Rlc, COOYOCOR10,
CH2OR10, CH2OCORlc, CH2OC(O)ORu
R9 is Rld, ORld, CH2-Hal, CH2ORld, CH2OC(O)ORπ, =CHRld
R10is Rle, CH=NORle, CN, COORle, CORle, CH2-Hal, CH2ORle,
CH2OCORle, CH2OC(O)ORπ, CH2OSO2CH3, CH2OSO2C6H4CH3
R11 is an OH-substituted alkyl group, an ether group or a cyclic ether;
R12 is lower alkyl substituted by Hal
"b" is a double bond or a single bond and wherein Y = (CH2)n n = 0 to 5
Rla"le are the same or different groups of R1 Hal is Cl, Br, I, F;
with the proviso that said compound is other than
3 β-3-hydroxyluρ-20(29)-en-28-oic acid;
3β-lup-20(29)-ene-3,28-diol;
3β-lup-20(29)-ene3,28-diol diacetate;
3β-3-(acetyloxy)luρ-20(29)-en-28-oic acid;
3β-3-(l-oxobutoxy)lup-20(29)-en-28-oic acid;
3 β-lup-20(29)-ene-3 ,28-diol 3-acetate;
3 β-lup-20(29)-ene-3 ,28-diol 28-acetate;
3β-3-hydroxylup-20(29)-en-28-oic acid methyl ester;
3β-3-(acetyloxy)lup-20(29)-en-28-oic acid methyl ester;
3β-3- hydroxylup-20(29)-en-28-oic acid ethyl ester;
3 β-3- hydroxylup-20(29)-en-28-oic acid butyl ester;
3 β-lupane-3 ,28-diol;
3β-3- hydroxylupan-28-oic acid;
3 β-3- hydroxylupan-28-oic acid methyl ester;
3β-3-(acetyloxy)lupan-28-oic acid methyl ester;
3β-3-(acetyloxy)lupan-28-oic acid;
3β-lupane-3,28-diol diacetate;
3β-lupane-3,28-diol dibutanoate;
3β-3-(3-methyl-l-oxobutoxy)lupan-28-oic acid;
3 β-3 -(2,2-dimethyl- 1 -oxopropoxy)lupan-28-oic acid;
3β-3,28-dimethoxylup-20(29)-ene;
3 β-3 ,28-dimethoxylupane;
3 β-28-methoxylupan-3 -ol;
3β-3-methoxylup-20(29)-en-28-oic acid;
3β-3-methoxylup-20(29)-en-28-oic acid methyl ester;
3α-3-methoxylup-20(29)-en-28-oic acid; or 3 -3-methoxylup-20(29)-en-28-oic acid methyl ester.
2. Use according to claim 1 wherein
X* is C=O, CHORla, CHOCORla, CHOCOY-Hal, CHOCOOR11;
X4 is C=0, CH2;
X5 is C=O, CH2, CHOCORlb;
R7 is COORlc, CH2ORlc, CH2OCORlc, COOYOCOR10,
CH2OSO2C6H4CH3, COHal, COOCOR10, CH2OCOORπ;
R9is Rld, =CHRld, CH2ORld, ORld;
R10 is Rle, CN, COORle, CH2ORle, CORle.
3. Use according to claim 1 or claim 2 wherein R11 is a diol, triol or polyol.
4. Use according to any one of claims 1 to 3 wherein
R1, R3"5 are methyl, R2 is H or methyl, and R1 is defined as below for the relevant group Rla"e; when X1 is:
-CHORla, Rla is H;
-CHCOY-Hal, Y is CH2 and Hal is Cl;
-CHOCORla, Rla is methyl;
-CHOCOOR11, R11 is glyceryl, 2,2-dimethyl-l,3-dioxolan-4-yl or pentaerythrityl; when X5 is CHOCORlb, Rlb is methyl; when R is:
-COORlc, Rlc is H or methyl;
-COOYOCORlc, Y is CH2 and Rlc is H;
-CH2ORlc, Rlc is H;
-CH2OCORlc, Rlc is methyl;
-COOCOR10, Rlc is methyl or butyl;
-COHal, Hal is Cl; -CH2OCOORπ, R11 is glyceryl, 2,2-dimethyl- 1, 3 -dioxolan-4-yl or pentaerythrityl; when R9 is =CHRld, CH2ORld or ORld, Rld is H, and when R9 is Rld, Rld is methyl; when R10 is Rle, Rle is methyl, and when R10 is COORle, CORle or CH2ORle, Rle is H.
5. Use according to any one of claims 1 to 4 wherein the compound of formula I is selected from the following table:
where: A is CHOC(O)OCH2CHOHCH2OH
B is CHOC(O)OCH2-2,2-dimefhyl-l,3-dioxolan-4-yl Ts is OSO2C6H4CH3. Use according to any preceding claim wherein said compound is selected from the following:
Use of a compound according to any one of claims 1 to 5 for treating a proliferative disorder.
Use of a compound of formula I, or a pharmaceutically acceptable salt thereof, in the preparation of medicament for use in the treatment of a proliferative disorder.
A compound of structural formula la, or a pharmaceutically acceptable salt thereof,
wherein:
X1 is CHOCOOR1 CHOC(O)ORla, CHOC(O)OR12, or CHOCOY- Hal
X4 is CH2, CH-Hal, C=O, CHORlb, CHOCORlb, CHOC(O)ORπ X5 is CH2, CH-Hal, C=O, CHORlb, CHOCORlb, CHOC(O)ORπ R1"5 are H or lower alkyl
R7 is COHal, C(O)OC(O)Rlc, COOYOCOR10, CH2OC(O)ORπ R9 is Rld, ORld, CH2-Hal, CH2ORld, CH2OC(O)ORπ, =CHRld R10is Rle, CH=NORle, CN, COORle, CORle, CH2-Hal, CH2ORle, CH2OCORle, CH2OC(O)ORn, CH2OSO2CH3, CH2OSO2C6H4CH3 R11 is an OH-substituted alkyl group, an ether group or a cyclic ether; R12 is lower alkyl substitued by Hal "b" is a double bond or a single bond and wherein Y = (CH2)n n = 0 to 5
Rla"leare the same or different groups of R1
Hal is Cl, Br, I, F.
10. A compound according to claim 9 wherein
X1 is C=O, CHORla, CHOCORla, CHOCOY-Hal, CHOCOOR11;
X4 is C=O, CH2;
X5 is C=O, CH2, CHOCORlb;
R7 is COORlc, CH2ORlc, CH2OCORlc, COOYOCOR10,
CH2OSO2C6H4CH3, COHal, COOCORlc, CH2OCOORπ;
R9is Rld, =CHRld, CH2ORld, ORld;
R10 is Rle, CN, COORle, CH2ORle, CORle.
11. A compound according to claim 9 or claim 10 wherein R11 is a diol, triol or polyol.
12. A compound according to any one of claims 9 to 11 wherein
R1, R3"5 are methyl, R2 is H or methyl, and R1 is defined as below for the relevant group Rla"e; when X1 is:
-CHORla, Rlais H;
-CHCOY-Hal, Y is CH2 and Hal is Cl;
-CHOCORla, Rla is methyl;
-CHOCOOR11, R11 is glyceryl, 2,2-dimethyl-l,3-dioxolan-4-yl or pentaerythrityl; when X5 is CHOCORlb, Rlb is methyl; when R7 is:
-COOR10, Rlc is H or methyl;
-COOYOCOR10, Y is CH2 and Rlois H;
-CH2OR10, Rlc is H;
-CH2OCORlc, Rlc is methyl;
-COOCOR10, R10 is methyl or butyl;
-COHal, Hal is Cl; -CH2OCOORπ, R11 is glyceryl, 2,2-dimethyl- l,3-dioxolan-4-yl or pentaerythrityl; when R9 is =CHRld, CH2ORld or ORld, Rld is H, and when R9 is Rld, Rld is methyl; when R10 is Rle, Rle is methyl, and when R10 is COORle, CORle or CH2ORle, Rle is H.
13. A compound selected from the following:
where: A is CHOC(O)OCH2CHOHCH2OH
B is CHOC(O)OCH2-2,2-dimethyl-l,3-dioxolan-4-yl Ts is OSO2C6H4CH3.
14. A process for preparing a compound of formula I, as defined in claim 1, wherein X4 is C=O and X5 is C=O or CH2,
comprising
(i) oxidising a compound of formula lb to form a compound of formula Ic; and (ii) where Rlc is H, esterifying with an appropriate group bearing the desired Rlc substituent; and optionally (iii) oxidising the product of step (i) or (ii) to form a compound of formula Id.
15. A process according to claim 14 wherein the compound of formula lb is oxidised to Ic by treating with ruthenium tetroxide.
16. A process according to claims 14 or 15 wherein the compound of formula Ic is oxidised to Id by treating with selenium dioxide.
17. A process for preparing a compound of formula I as defined in claim 1, wherein X4 and X5 are CH2,
comprising oxidising a compound of formula le to form a compound of formula If.
18. A process according to claim 17 wherein the compound of formula le is oxidised to If by treating with selenium dioxide.
19. A process for preparing a compound of formula I, as defined in claim 1, wherein R1"5 are methyl,
ig Ih
X4 = C=θ, X5 = CH2 C=θ, Xs = CH2 = C=θ, Xs = CH2 R9 = CH3, R10 = CH3 R3 = CH3, R10 = CH3 = CH3, R — CH3 R7 = COOCH3 = COOCH3 = COOCH3 b is double bond b Is double bond b Is double bond
X4 = C=θ, X5 = CH2 = C=θ, X5 = CH. X4 = C=θ, Xs = CH2 R9 = CH3, R 0 = CH3 ■ CH3, R CH, : CH3, R — CH3 R7 = COOH = COOH = COOH b is double bond b is double bond b is double bond
X4 = C=θ, X5 = C=θ = c=o, x3 = c=o X = c=o, xb = c=o R9 = CH3, R10 = CH3 = CH3, R10 = CH3 = CH3, R = CH3 R7 = COOCH3 = COOCH3 = C00CH3 b is double bond is double bond b is double bond
X — Cπ , X — CH CH , X = CH X4 = CH2, X& = CH2 R9 = =CH2, R10= CH3 ?s = CH2l R =3 =CH2, R10 = CH,
37 = R7 = COOH = COOH COOH b is single bond b is single bond b is single bond
X — CH , X ~~ CH2 = CH2, X6 = CH, - CH2, X — CH R9 = =CH2, R10 = CH3 = — CH2l R — CH3 = =CH2, R = CH3 R = COOCH3 = COOCH3 = COOCH3 b is single bond b is single bond b is single bond
X = CH2, X5 = CH2 X — CH2, X — CH X = CH2, X — CH R9 = =CH2, R10 = CH3 R9 = =CH2l R10 = CH3 = — CH2, R = CH3 R7 = COOCH2OCOCH3 = COOCH2OCOCH3 j = COOCH2OCOCH3 b is single bond b is single bond b is single bond
said process comprising the steps of:
(i) reacting a compound of formula Ig with chloroformate of solketal to form a compound of formula Ih; (ii) deprotecting said compound of formula Ih to form a compound of formula Ii.
20. A process for preparing a compound of formula I, as defined in claim 1, wherein R^R5 are methyl, X5 is CH2, X1 is CHOAc or C=O, X4 is CH2 or C=O, R9 is =CH2 or CH3, R10 is CH3 or COH and R7 is COOH or COF,
X1 = CHOAc, X4 = CH2 X1 = CHOAc, X4 = CH2 R9 = =CH2, R™ = CH3 R9 = =CH2, R10 = CH3 b is single bond b is single bond or or
X1 = CHOAc, X4 = CH2 X1 = CHOAc, X" = CH2 Ra = CH3, R10 = CH3 R9 = CH3, R10 = CH3 b is single bond b is single bond or or
X< = C=0, X" = CH2 X1 = C=0, X* = CH2 R9 = =CH2, R10 = CH3 R9 = =CH2, R10 = CH3 b is single bond b is single bond or or
Xi = CHOAc, X* = CH2 Xi = CHOAc, X" = CH2 R9 = =CH2, 1° = COH R9 = =CH2, R10 = COH b is single bond b is single bond or
Xi = CHOAc, X4 = C=0 X1 = CHOAc, X4 = C=0 R9, R1° = CH3 R9, R10 = CH3 b is double bond b is double bond
said process comprising reacting a compound of formula of Ij with diethylaminosulphur trifluoride to form a compound of formula Ik.
EP01936618A 2000-05-23 2001-05-23 Triterpenoid derivatives Withdrawn EP1292562A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GB0012528 2000-05-23
GBGB0012528.6A GB0012528D0 (en) 2000-05-23 2000-05-23 Triterpenoid derivatives
GB0012823A GB2362649A (en) 2000-05-25 2000-05-25 Triterpenoid derivatives
GB0012823 2000-05-25
PCT/GB2001/002309 WO2001090046A1 (en) 2000-05-23 2001-05-23 Triterpenoid derivatives

Publications (1)

Publication Number Publication Date
EP1292562A1 true EP1292562A1 (en) 2003-03-19

Family

ID=26244332

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01936618A Withdrawn EP1292562A1 (en) 2000-05-23 2001-05-23 Triterpenoid derivatives

Country Status (3)

Country Link
EP (1) EP1292562A1 (en)
AU (1) AU2001262489A1 (en)
WO (1) WO2001090046A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2178376A4 (en) * 2007-08-03 2011-12-14 Advanced Life Sciences Inc Lupane-type triterpenoids modified at 30-position and analogues thereof
CA2711424A1 (en) * 2008-01-03 2009-07-09 Virochem Pharma Inc. Novel lupane derivatives
JP5320530B2 (en) * 2008-02-14 2013-10-23 秋田県 Triterpene compound useful as anticancer agent and anticancer composition using the triterpene compound
CZ2008723A3 (en) 2008-11-13 2010-01-13 Univerzita Palackého v Olomouci 2-Deoxyglycosides of triterpenoids, process of their preparation and their use as medicament
JO3387B1 (en) 2011-12-16 2019-03-13 Glaxosmithkline Llc Derivatives of betulin
RU2551647C2 (en) * 2012-11-12 2015-05-27 Федеральное государственное бюджетное учреждение науки Институт нефтехимии и катализа Российской академии наук Triphenylphosphonium salts of lupane triterpernoids, method for producing and using as anticancer agents
CA2893959A1 (en) 2012-12-14 2014-06-19 Shenshen Cai Pharmaceutical compositions
BR112017006005A2 (en) 2014-09-26 2017-12-19 Glaxosmithkline Ip No 2 Ltd pharmaceutical composition, and method of preventing or treating a hiv infection.

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1415601A (en) * 1973-03-14 1975-11-26 Biorex Laboratories Ltd Dihydrobetulinic acid derivatives
US5962527A (en) * 1995-03-21 1999-10-05 The Board Of Trustees Of The University Of Illinois Method and composition for treating cancers
EP0943620B1 (en) * 1998-03-18 2005-11-16 Dabur Pharma Ltd. Betulinic acid derivatives for inhibiting cancer growth
US6369109B1 (en) * 1998-10-28 2002-04-09 Deutsches Krebsforschungszentrum Stiftung Des Offentlichen Rechts Betulinic acid and derivatives thereof useful for the treatment of neuroectodermal tumor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0190046A1 *

Also Published As

Publication number Publication date
WO2001090046A1 (en) 2001-11-29
AU2001262489A1 (en) 2001-12-03

Similar Documents

Publication Publication Date Title
US7749988B2 (en) Triterpenoid derivatives
CN108348530B (en) Fanisole X receptor modulators
AU2001284946B2 (en) Prodrugs of betulinic acid derivatives for the treatment of cancer and HIV
AU2001284946A1 (en) Prodrugs of betulinic acid derivatives for the treatment of cancer and HIV
Borkova et al. Synthesis of cytotoxic 2, 2-difluoroderivatives of dihydrobetulinic acid and allobetulin and study of their impact on cancer cells
WO2001090046A1 (en) Triterpenoid derivatives
EP3013847B1 (en) Therapeutically active estratrienthiazole derivatives as inhibitors of 17.beta.-hydroxy-steroid dehydrogenase, type 1
JP3913475B2 (en) Steroid sulfatase inhibitor and method for producing and using the inhibitor
US7858606B2 (en) Triterpenoid derivatives
JPH08269065A (en) Pyripyropnene derivative
US7732493B2 (en) 2-substituted D-homo-estra-1,3,5(10)-trienes as inhibitors of 17β-hydroxy steroid dehydrogenase type 1
Huang et al. C21-steroidal pregnane sapogenins and their derivatives as anti-inflammatory agents
EP1289977B1 (en) Triterpenoid derivatives and their use as antiproliferative agents
Li et al. Synthesis and discovery of mitochondria-targeting oleanolic acid derivatives for potential PI3K inhibition
Hou et al. Synthesis of oridonin derivatives via mizoroki-heck reaction and click chemistry for cytotoxic activity
Siwach et al. 1, 2, 3‐Triazole‐based esters and carboxylic acids as nonclassical carbonic anhydrase inhibitors capable of cathepsin B inhibition
Rárová et al. Effect of modification of betulinic acid at the C3-carbon atom of homolupane triterpenoids on the antiproliferative activity in vitro
ES2312965T3 (en) SULFAMATES OF D-HOMOESTRA-1,3,5 (10) -TRIEN-3-IL-2-SUBSTITUTED WITH ANTITUMORAL EFFECTS.
GB2362649A (en) Triterpenoid derivatives
US7151116B2 (en) Apoptolidin analogs and derivatives for inducing apoptosis in transformed cells
RU2801166C1 (en) [n&#39;-(iso)quinolylmethylene]hydrazides of 3-methoxy-13,17-secoestra-1,3,5(10)-trien-17-oic acid
GB2362650A (en) Triterpenoid derivatives
CN115785189B (en) 5 alpha, 8 alpha-peroxosterol-17-phenylthiazole derivative and synthetic method and application thereof
ZhenYu et al. Synthesis and Anti-Tumor Activity of Oleanolic Acid Derivatives
CN116375582A (en) Isosteviol derivative and application thereof

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20021223

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20070619

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20161201