GB2362649A - Triterpenoid derivatives - Google Patents

Triterpenoid derivatives Download PDF

Info

Publication number
GB2362649A
GB2362649A GB0012823A GB0012823A GB2362649A GB 2362649 A GB2362649 A GB 2362649A GB 0012823 A GB0012823 A GB 0012823A GB 0012823 A GB0012823 A GB 0012823A GB 2362649 A GB2362649 A GB 2362649A
Authority
GB
United Kingdom
Prior art keywords
compound
double
formula
choac
cooh
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Withdrawn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
GB0012823A
Other versions
GB0012823D0 (en
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
Application filed by Karlova Univerzita v Praze, Univerzita Palackeho V Olomouci filed Critical Karlova Univerzita v Praze
Priority to GB0012823A priority Critical patent/GB2362649A/en
Publication of GB0012823D0 publication Critical patent/GB0012823D0/en
Priority to PCT/GB2001/002309 priority patent/WO2001090046A1/en
Priority to EP01936618A priority patent/EP1292562A1/en
Priority to AU2001262489A priority patent/AU2001262489A1/en
Publication of GB2362649A publication Critical patent/GB2362649A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Emergency Medicine (AREA)
  • Epidemiology (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 thereof in therapy, particularly for treating a patient suffering from leukaemia, cancer or other proliferative disorder. A further embodiment relates to the use of a compound of formula I in an assay for detecting the phosphorylation state of cellular substrates. The present invention also relates to certain novel compounds of formula I, and the chemical synthesis thereof. <EMI ID=1.1 HE=51 WI=64 LX=792 LY=1162 TI=CF> <PC>wherein the symbols are as defined in the specification.

Description

2362649 TRITERPENOID 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 1, flavopiridol, bohernin, 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 GI/S transition. Unlike normal cells, tumour cells do not arrest in G1, and since they become independent of growth factors, they pass the G1 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 CM 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 Sphase transcription factors such as DP-1 and E2F. However, following phosphorylation by G1IS cyclin-dependent 2 kinases (CDKs) (C13K4/cyclin DI-D3, CM6/cyclin DI-D3, CM2/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 CM2/cyclin E complexes. Thus, hyperphosphorylation of the pRb protein plays a key role in the molecular pathology of cancer cells with altered CM activity.
The present invention relates to the use of triterpenoid compounds derived fi7om 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.
--J."
H H R HO R = CH20R 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, 23 32 (1922), Schulze H. et al, Acta Chem. Scand., B 29, 13 9 (1975), Suokas E. et al; Collect. Czech. Chem. Commun. 56, 2936 (1991), Sejbal I et al, Collect.
3 Czech. Chem. Commun. 64, 329 (1999), Klinotovi 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, R R9 v X5 R3 R 4 R7 H xl R5 H R2 R, 1 wherein:
X' is C=Q CHOW', CHOCOW'. or CHOCOY-Hal X4 is CH2, C=0 X5 is CH 2, C=O, CHOCOR1b R1-5 are H or lower alkyl IC is C0OW', CO0Y0COW', CH2OW', CH20COW' R is R 1d 0Rld, CH 2 0Rld =CIHR 1d R10 is Rle, CN, C0OW', COW', CH2OR le W' is a double bond or a single bond and wherein Y (CH2)n n 0 to 5 W"' are the same or different groups of R' Hal is Cl, Br, 1, F.
1 4 In a preferred aspect, the invention relates to the use of a compound of formula 1, 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, R10 R9 -_ X4 H X5 R 3 R4 R7 X, -- H R5 H R2 R, wherein:
la X'is C=O, CHOR". CHOCOW', or CHOCOY-Hal X4 is CH2, C=0 X5 is CH2, C=O, CHOCOR 1b R1-5 are H or lower alkyl R7 is COOR", CO0Y0COC, CH2OW', CH2OCOW' R is R 1d, OR W5 CH2OR 1d, =CM1d R10 is C, CN, COOR", COR', CH2OW' "b" is a double bond or a single bond and wherein Y (CH2)n n 0 to 5 W"' are the same or different groups of R' Hal is Cl, Br, 1, F.
1 with the proviso that:
(i) when X' is CHOAc, W-, R and R10 are all Me, - when X4 is C=Q X is CH2 and "b" is a double bond, R is not COOH, CH20Ac or COOMe; and - when X4 and X5 are C=0 and "b" is a double bond, or when X is CHOAc and "b" is a single bond, R7 is not CH20Ac; - when X4 and X-5 are CH2 and "b" is a double bond, R7 is not CH20Ac, C02Me or H; (ii) when X1 is CHOH or CHOAc, X4 and X' are CH2, "b" is a single bond, R1-5 are Me, - IC is not COOH when R? is CH2, RO is CHO; and - IC is not CH20Ac or C02Me, when R!, R10 are Me; (iii) when X' is C=Q X4and X are CH2, "b" is a single bond, C', R and W'D are Me, R7 is not C001t Me or CH20Ac; (iv) when X' is CHOH, X4 and X5 are CH2, "b" is a double bond, W-5, R, R10 are Me, R7 is not COOH.
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 X', X, X5 and W-5, IC, R-10 of formula 1, the following options are preferred; Preferably, R', C' are methyl, R is H or methyl, and R' is defined as below for the relevant group R la-e 1 6 More preferably, R-5 are all methyl. Preferably, when X' is: -CHOR la, W' is preferably H; -CHCOY-Hal, Y is preferably CH2 and Hal is preferably Cl; Preferably, when X5 is CHOCOR 1b, R 1b is preferably methyl. Preferably, when R is: -CHOOR", R1c is preferably H or methyl, -CO0Y0COW', Y is preferably CH2 and W' is preferably H; -CH2OW', W' is preferably H; -CH20COR1c, W' is preferably methyl. Preferably, when W is =C]HRId, CH2OR Id or OR"', R Id is preferably R and when R is R Id, R Id is preferably methyl. Preferably, when R10 is R", C is preferably methyl, and when R10 is COORle CORle or CH20Rle, W' is preferably 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.
7 Table 1 (Compounds of general structure 1).
No. 1 xl X 1 b W"' 1 R' 1 FC R- 11.1 CHOAc C=0 CH2 double Me COOH Me Me 11.2 CHOAc C=0 C=0 double Me CH20Ac Me Me 11.3 CHOAc C=0 CH2 double Me CH20Ac Me Me 11.4 CHOAc C=0 C=0 double Me COOMC Me Me 11.5 CHOCOCH2C1 C=0 CH2 double Me CH20Ac Me Me 11.6 CHOCOCH251 C=0 C=0 double Me CH20Ac Me Me 11.7 CHOH C=0 C=0 double Me CH20Ac Me Me 11.8 CHOH C=0 C=0 double Me CH20H Me Me 11.9 CHOAc C=0 CH2 double Me COO H20-COU' Me Me 11.10 CHOAc C=0 CH2 double Me COOMe Me Me 11.11 CHOAc C=0 C=0 double Me COOCH20C013u Me Me 11.12 CHOAc CH2 CH2 single Me COOH CH2 CN 11.13 CHOAc CH2 CH2 single Me COOH CH2 COOH 11.14 CHOH CH2 CH2 single Me COOH CH2 COH 11.15 CHOAc CH2 CH2 single Me COOH CH2 CH20H 11.16 CHOAc C=0 CHOAc double Me CH20Ac Me Me 11.17 C=0 CH2 CH2 single Me, CH20Ac Me Me R 2: H) 11.18 CHOAc CH2 CH2 single Me COOH CH2 COH 11.19 CHOH CH2 CH2 single Me COOH (20-R)-OH Me 11.20 CHOH CH2 CH2 single Me COOH (20-S)-OH Me 11.21 CHOH CH2 CH2 single Me COOH (20-R)-CH20H Me 11.22 CHOH CH2 CH2 single Me COOH (20-S)-CH20H Me 11.23 CHOAc C=0 C=0 double Me COOCH20COMe Me Me 11.24 CHOH CH2 CH2 single Me COOCH2OCOU' CH2 COH 11 - 25 CHOH CH2 CH2 single Me COOCH20COMe CH2 c 6 CHOH C=0 C=0 double Me 00Me Me Me LIS CHOH C=0 CH2 double Me COOCH20COBUI Me Me 11.27 1. 27 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 Ia.
The most preferred compounds of the second aspect are those in Table 1 not marked, i.e. compounds HA-11.9, H. 11-11. 13, H. 15-11. 17, 11. 19-H. 27.
1 8 In respect of the invention as a whole, it is preferable that the proliferative disorder is cancer or leukaemia. More preferably, the cancer or leukaemia is p53, Rb, hormone and multidrug resistance independent.
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 1 1 9 proliferative cells comprising administering a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salts or esters thereof 1; 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 10 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 1 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 1.
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 30 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 CM, 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/n-A penicillin and piglml 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 ig/ml of leupeptin, 10 ggImI of aprotinin, 10 Rg/mI of soybean trypsin inhibitor, 100 Rmol of benzamide, 1 mM of sodium vanadate, 1 mM of NaF, 1 mM of phenylphosphate) and boiled immediately.
1 11 Total cellular proteins (100 gg/well) are separated on SDS-PAGE electrophoresis, blotted on polyvinyldifluoride membranes and total Rb protein, including proteolytic ftagment(s) detected using a pRb monoclonal antibody (Oncogene, Germany, Rb(Ab-5), Cat# OP66 Rev 02-Sept- 96 EB, Clone 5 LM95. 1) and visualized by chemiluminiscence (ECL-Western 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 I 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-C4)-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methaneor ptoluene sulfonic acid.
12 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, flamaric, 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 (Cl-C4)-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 Ia. 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.
1 1 13 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 Ia 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 10 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 14 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 I 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 15 administered to the patient for the treatment of malignancy.
The invention finiher 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 1, as defined above, wherein X4 is C=0 and X5 is C=0 or CH2, 0 H FR3 R4 00OR1c H RS X, f". F ' ' _ H R2 R, 0 H 1 1 R,R,COOF' H 1 1 X, - PR4R, H R2 R, 0 H 1 0 R3 R4 c cc HR, X ' H R 2 R, 1b [c 1d comprising 16 (i) oxidising a compound of formula 1b to form a compound of formula lc.
and (ii) where R1c is R esterifying with an appropriate group bearing the desired W' 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 lc 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 1 as defined above, wherein X4and X5 are CH2, -J., H R3 R4 COOFc r""1 1 H R5 X, H R 2 R, H 0 -Y H R3 R4 coofc X, H R5 H R 2 R, ilia le If 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.
17 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 X' is CHOAc, X4 is C=Q X5 is CH2 or C=Q W-5,9,10 are methyl, and R7 is CH20Ac, COOMe or COOCH20COC(CH3)3.
c0 0COR1c AcO a [ R10 = CH3 R=H c0 0 0 b COOR1c f COOR1c AcO AcO RIc = H c RIc CH3 R1c = CH3 e R1c = CH20COC(CH3)3 d R1c = CH20COC(CH3)3 R1c = CH20AC F- Rlc=CH20AC-'- 0 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 X' is CHOAc, X4,5 are CH2, R1-5 are methyl, R7 is COOR R is CH2, and R10 is COH.
_,j, COOH HO 1 - 0 1 H 0 a OOH HO 18 Conditions: a, oxidation (e.g. with selenium dioxide).
This invention is further illustrated by the following examples, which should not 5 be construed as ffirther limiting.
19 EXAMPLES
General The chemical shift values (5-scale, ppm) and coupling constants Q, Hz) in the 1H and 13 C NMR spectra were obtained using a Varian =Y-INOVA 400 FT spectrometer (H at 400 NfHz and 13C at 100.6 MHz) in deuterochloroform with tetramethylsilane (for 1H NMR data 5 = 0 ppm) as an internal standard. For the 13C NMR data 5(CDC13)=77.00 ppm. The value for a multiplet, 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 (EM) were measured on an INCOS 50 instrument. Ionising electron energy 75 eV, ion source temperature 150 'C. EIMS was used to determine molecular weights, M+ corresponding to the molecular ion.
Ether is diethylether. THIF 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.
EXAWLE 1 Lup-20(22)-ene-30,28-diyl diacetate Crude betuline (500 g) was dissolved in a mixture of 250 mi pyridine and 250 nil 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-30,28-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 re-crystallization from chloroforni/methanol, the yield of pure compound was 239 g, mp. 222 - 223 'C, [OCID+22' (c 0.4; CHC13). [Schulze H., Pieroh K.: Ber. Dtsch. Chem. Ges. 55, 2332 (1922)].
The 'H 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 R 2.07 s, 3 H (2x 0Ac); 2.44 ddd, 1 H (F= 11.4, F' = 10.9, J... = 0.7, H19); 3.85 d, 1 H Q = 11. 1, H-28a); 4.25 dd, 111, (F = 11. 1, F' = 1.4, H-28b); 4.47 m, 1 H (H 3a); 4.59 ni, 1 H (E J = 3.4, H-29E); 4.69 m, 1 H (E J = 2. 1, H-29Z).
EXAWLE 2 Lup-18-ene-30.28-diyl diacetate A solution of hydrogen bromide in acetic acid (38 %, 1.4 1) was added to a solution of lup-20(29)-ene-30,28-diyl diacetate (100 g, 190 mmol) in a mixture of benzene, acetic acid and acetic anhydride (1 1: 0.5 1 - 50 M1). The reaction mixture was refluxed until the reaction was completed (TLC was developed in hexane/ether mixture). After cooling, the reaction mixture was poured into ice cold water (M) and extracted with benzene (3 x 0.5 1). The combined organic phases were washed with NaHC03 aqueous solution, NaHS03 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 SoxhIet 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 %), 1 21 mp. 215-216 'C, [a]D +151 (c 0.45; CHG3). [Suokas E, Hase T.: Acta Chem. Scand., B 29, 139 (1975)].
The 'H 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 Q = 6.8), 0.99 d, 3 H Q = 6. 8), 1.06 s, 7 - 2.25 m, 2 H (E J - 15); 2.43 x 3H (7 x CH3); 2.04 s, 3 R 2.05 s, 3 H (2x 0Ac), m, 1 H (E J - 15); 3.14 sept, 1 H (J = 7, H-20); 3.98 d, 1 H Q = 10.8, H- 28a); 4.03 d, 1 H Q = 10.8, H-28b); 4.49 m, 1 H (11-3 cc).
EXAMPLE 3 2 1 -oxo-lup- 1 8-ene-3 0.28-diyl diacet Lup-18-ene-30,28-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 nil).
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 finther purification (see Example 4). Pure title compound has m.p. 205 - 206 OC, [(X1D -35' (c 0.49; CHC13). Another way to the title compound is described in Sejbal L, Klinot L, Budeginsk M, Protiva 1: Collect. Czech. Chem.
Commun. 56,2936 (1991).
The 'H 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 Q = 7.1), 1.21 d Q = 7. 1), 7 x 3 H (7x CH3); 2.00 s, 3 a 2.05 s, 3 H (2x 0Ac); 2.39 d, 1 H Q= 18.5, H-22); 2. 87 dd, 1 H Q '= 11. 9, F' = 4. 1, H- 13 0); 3.18 sept, 1 H Q = 6.6, H-20); 4. 06 d, 1 H Q = 10.9, H-28a); 4.34 d, 1 H Q = 10.9, H-28b); 4.49 m, 1 H Q - 7, H-3a).
22 The following compounds were prepared by the above-mentioned procedure:
(pivaloyloxy)methyl 30,28-diacetoxy-18-oxo-19,20,21,29,30-pentanorlupan22- oate acetoxyinethyl 30,28-diacetoxy-18-oxo-19,20,21,29,30-pentanorlupan- 22- oate EXAMPLE 4
21,22-dioxolup-18-ene-30.28-diyI diacetat A solution of crude 21-oxolup-18-ene-30,28-diyl diacetate (40g; 74 mmol; containing about 85 % of 2 1 -oxo-lup- 1 8-ene-3 0,28-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 redorange precipitate was filtered off under reduced pressure, carefully washed with water and dried in the air. Dry crude 21,22-dioxolup-18-ene-3p,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, [U.]D -127' (c 0. 32; CHC13). Another way to the title compound is described in Klinotovi E, CermAovA 1, Rejzek M., Krecek V., Sejbal L, Olgovsk R, Klinot L Collect. Czech. Chem. Commun. 64, 329 (1999).
The 'H 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 Q = 7.2), 1.26 d Q = 7.2), 7 x 3 H (7x CH3); 1.93 s, 3 R 2.06 s, 3 H (2x 0Ac); 3.12 dd, 1 H Q '= 12. 5, F' = 3.8, 23 H-130); 3.36 sept, 1 H Q= 7.0, H-20); 4.02d, 1 H Q= 11.1,H-28a); 4.49 dd, 1 H (F = 10.2, J- = 6.0, H-3a); 4.84 d, 1 H Q = 11. 1, H-28b).
The following compounds were prepared by the above-mentioned procedure:
methyl 30-acctoxy-21,22-dioxolup-18-en-28-oate acetoxymethyl 3p-acctoxy-21,22-dioxolup-18-en-28-oate (pivaloyloxy)methyl 30-acetoxy-21,22-dioxolup-18-en-28-oate acetoxymethyl-3 0,28-diacetoxy- 1 8-oxo- 19,20,21,29,3 0-pentanorlup- 1 2- en-22oate (pivaloyloxy)methyl 30,28-diacetoxy-lg-oxo-19,20,21,29,30-pentanorlup- 12en-22-oate 30-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)1. acetoxymethyl 30-hydroxy-30-oxolup-20(29)-en-28-oate 15 (pivaloyloxy)methyl 30-hydroxy-30-oxolup-20(29)-en-28-oate EXAWLE 5 28-hydroU-2 I -oxolup- I 8-en-3 0-yl acetat A solution of 21-oxolup-18-ene-30,28-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 (M) 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, [CCID 69' (c 0.34; CHG3). [MinotovA E, CermAkovi J, Rejzek M, Krecek V, Sejbal J., Olgovsk P, 30 Klinot L Collect. Czech. Chem. Commun. 64, 329 (1999)1.
24 The 'H 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 Q = 6.9), 1.21 d Q = 6.9), 7 x 3 H (7x CH3); 1.92 d, 1 H Q = 18.6, H-22a); 2.05 s, 3 H (0Ac); 2.44 d, 1 H Q= 18. 6, H-22b); 2.78 dd, 1 H Q '= 12.5, J---= 3.4, H- 13 p); 3.19 sept., 1 H Q = 6.9, H-20); 3.67 d, 1 H Q = 10.7, H-28a); 3.72 d, 1 H Q = 10.7, H-28b); 4.49 dd, 1 H (J'= 11,0, J- = 5.5, H-3(x).
EXAWLE: 6 BIOLOGICAL ACTIVITY OF BETULR-'MS In vitro gAotoxic 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 [Hajd6ch K NfihAl V, Nfinarik J, Fiber E, 9afirovi M, Weigl E, Antilek 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/mI penicillin, 100 pg/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 (80PI) into 96/well microtiter plates. Inoculates were allowed a pre-incubation period of 24 hours at 37'C and 5% C02 for stabilisation. Four-fold dilutions of the intended test concentration were added at time zero in 20d 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 pK 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% C02 atmosphere and 100% humidity. At the end of the incubation period, the cells were assayed by using the NITT 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 100gl/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 NIF(LTK). The tumour cell survival (TCS) was calculated using the following equitation: TCS=(ODd,.g exposed wen / mean ODcontrol wefls) X 100%. The TC S5o value, the drug concentration lethal to 5 0% of the tumour cells, was calculated from the obtained dose response curves.
26 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 the most effective betulinines against a panel of different (non)malignant cell lines.
Compound (TCS5o[tLM]) Cell Line Description Betulinic 11.4 11.7 11.14 11.26 acid B16 mouse melanoma 36 35 9.3 B16F mouse melanoma, metastatic 4.6 68 12 SW620 human colon cancer, metastasis 250 5.9 5-9 U87MG human glioblastorna 250 24 8.4 HepG2 human hepatocellular carcinoma 3.6 5.1 7.1 A549 human lung adenocarcinoma 236 6.7 3.3 MCF-7 human breast cancer, estrogen 194 13 4.3 dependent, p53+/+, Rb +/+ U20S human osteosarcoma, p53+/-, Rb +/- 250 4.3 3.9 Saos2 human rhabdomyosarcoma, p53-/-, Rb-/- 250 7.9 4.8 BT549 human breast cancer, p53muttmut 250 15 6.3 MDA-MB-238 human breast cancer, estrogen 195 3.3 2.4 independent, p53muttmut LNCaP human prostate cancer, androgen 244 3.3 3.6 de endent DU145 human prostate cancer, androgen 241 2.0 2.8 independent, Rb-/ HT-29 human colon cancer 250 5.6 4.7 OVCAR-3 human ovarian cancer 164 4.7 2.6 Gaco-2 human colon cancer 20 8.1 7.7 MEL-3 human melanoma 2.7 5.9 3.7 Lymphocytes human normal lymphocytes 250 85 25 NlH3T3 mouse immortalised fibroblasts 250 16 27 Table 2 continued Compound (TCS5o[pM]) Cell Line Description Betulinic 11.4 11.7 11.14 11.26 acid K562-CdA human promyelocytic leukemia, 250 5.2 2.1 cladrubin resistant K562-GEM human promyelocytic leukemia, 101 5.0 2.4 gerneitabin resistant K562-ARA-C human promyelocytic leukemia, 250 6.4 1.6 cytarabin resistant K562-FLUD human promyelocytic leukemia, 250 4.9 1.3 fludarabin resistant CEM human T-lymphoblastic leukemia 250 7.2 6.3 3.0 1.1 CEM-DNR human T-lymphoblastic leukemia, 250 10 1.6 I/C2 daunorubicin resistant CEM-DNR bulk human T-lymphoblastic leukemia, 250 5.2 3.3 daunorubicin resistant CEM-VCR 11F3 human T-lymphoblastic leukemia, 19 38 5.9 vincristin resistant CEM-VCR 3/D5 human T-lymphoblastic leukemia, 24 13 7.8 vincristin resistant CEM-VCR bulk human T-lymphoblastic leukemia, 69 28 7.0 vincristin resistant In contrast to betulinic acid, which is reported to be an agent selective for 5 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.
1

Claims (24)

Z9 CLAIMS Use of a compound of formula 1, or a pharmaceutically acceptable salt thereof, in therapy, R10 R9 X.4 H X5 R 4 R7 1H f R ,5 R21 RI- wherein:
1 X' is C=Q CHOR", CHOCOW', or CHOCOY-Hal X4 is CH2, C=0 X5 is CH 2, C=O, CHOCOR1b W" are H or lower alkyl R7 is C0OW', CO0Y0COW', CH2OW', CH20COW' R is R Id 7 0Rld, CH 2 0Rld. =CM1d R10 is C, CN, COORle, COW', CH2OW' W' is a double bond or a single bond and wherein Y (CH2)n n 0 to 5 W"' are the same or different groups of R' Hal is Cl, Br, 1, F.
2. Use of a compound according to claim 1 wherein R', R3-' are methyl, R is H or methyl, and R' is defined as below for the relevant group W; when X' is:
-CHOR la, R la is H; -CHCOY-Hal, Y is CH2 and Hal is Cl; when X5 is CHOCORib, R"> is methyl; when R is:
lc -CHOOR, W' is H or methyl; -CO0Y0COW', Y is CH2 and W' is H; -CH2OW', W' is H; -CH20COW', W' is methyl; when R is =CHRId, CH20Rld or OR 1d, R Id is R and when R is 1d 1d is M h 1.
R R et y when R10 is C, W' is methyl, and when W0 is COOC, COW' or CH2OR", W' is H.
31
3. Use of a compound according to claims 1 or 2 wherein the compound of formula 1 is selected from the following table:
No. 1 xl)C 1 X' 1 b 1 R 1 Rf 1 W 1 R 11.1 CHOAc C=0 CH2 double Me COOH Me Me 11.2 CHOAc C=0 C=0 double Me CH20Ac Me Me 11.3 CHOAc C=0 CH2 double Me CH20Ac Me Me 11.
4 CHOAc C=0 C=0 double Me COOMe Me Me 11.
5 CHOCOCH2C1 C'=0 CH2 double Me CH20Ac Me Me 11.6 CHOCOCH2C1 C=0 C=0 double Me CH20Ac Me Me 11.7 CHOH C=0 C=0 double Me CH20Ac Me Me 11.8 CHOH C=0 C=0 double Me CH20H Me Me 11.9 CHOAc C=0 CH2 double Me CO0CH20-COU' Me Me 11.10 CHOAc C=0 CH2 double Me COOMe Me Me 11.11 CHOAc C=0 C=0 double Me CO0CH2OCUBE Me Me 11.12 CHOAc CH2 CH2 single Me COOH CH2 CN 11.13 CHOAc CH2 CH2 single Me COOH CH2 COOH 11.14 CHOH CH2 CH2 single Me COOH CH2 COH 11.15 CHOAc CH2 CH2 single Me COOH CH2 CH20H 11.16 CHOAc C=0 CHOAc double Me CH20AC Me Me 11.17 C=0 CH2 CH2 single Me, CH20Ac Me Me (R2: H) 11.18 CHOAc CH2 CH2 single Me COOH CH2 COH 11.19 CHOH CH2 CH2 single Me COOH (20-R)-01-1 Me 11.20 CHOH CH2 CH2 single Me COOH (20-S)-OH Me 11.21 CHOH CH2 CH2 single Me COOH (20-R)-CH201-1 Me 11.22 CHOH CH2 CH2 single Me COOH (20-S)-CH201-1 Me 11.23 CHOAc C=0 C=0 double Me COOCH20COMe Me Me 11.24 CHOH CH2 CH2 single Me CO0CH20COU' CH2 COH 11.25 CHOH CH2 CH2 single Me COOCH20COMe CH2 COH 11.26 CHOH C=0 C=0 double Me COOMe Me 11.27 CHOH C=0 CH2 double Me COOCH20COBUI Me 4. Use of a compound according to claims 1 to 3 for treat ing a proliferative disorder.
32 5. Use of a compound according to claim 4 wherein the proliferative disorder is cancer.
6. Use of a compound according to claim 4 or 5 wherein the proliferative disorder is leukaemia.
Use of a compound according to claims 4 to 6 wherein the proliferative disorder is multidrug resistant.
8. Use of a compound according to claims 4 to 7 wherein the proliferative disorder is independent of p53.
9. Use of a compound according to claims 4 to 8 wherein the proliferative disorder is independent of Rb.
10. Use of a compound according to claims 4 to 9 wherein the proliferative disorder is independent of hormones.
11. Use of a compound according to any preceding claim wherein the compound of formula 1, or pharmaceutically acceptable salt thereof, effects a change in the Rb protein phosphorylation state.
12. Use of a compound according to any preceding claim for inducing programmed cell death in proliferative cells.
13. Use of a compound of formula 1, or a pharmaceutically acceptable salt thereof, in the preparation of medicament for use in the treatment of a proliferative disorder as defined in any of claims 4 to 10.
33
14. Use according to claim 13 comprising use of a compound of formula I in an assay for determining the phosphorylation state of cellular substrates.
15.
Use according to claim 14 wherein said assay is capable of identifying candidate compounds that influence substrate phosphorylation.
16. Use according to claim 14 or 15 wherein the cellular substrate is Rb protein.
17. A compound of structural formula Ia, or a pharmaceutically acceptable salt thereof, R10 R9 --J\\- X4 H X5 R3 r'4 R7 la wherein: X' is C=Q CHOR", CHOCOW', or CHOCOY-Hal X4 is CH2, C=0 X5 is CH 2, C=O, CHOCOR1b R1-5 are H or lower alkyl R7 is CO0C, CO0Y0CORle. CH20Rle, CH20CORle R is R ld, 0Rld, CH 2 0Rld. =CHR1d R10 is R", CN, C0OW'. COR', CH2ORIO 34 'W' is a double bond or a single bond and wherein Y (CH2)n n 0 to 5 W"' are the same or different groups of R' Hal is Cl, Br, I, F.
with the proviso that:
(i) when X' is CHOAc, R1-5, R and R10 are all Me, - when X' is C=O, X' is CH2 and 'W' is a double bond, R7 is not ' and COOH, CH20Ac or COOMe. when X4 and X' are C=0 and "b" is a double bond, or when X5 is CHOAc and "b" is a single bond, R7 is not CH20Ac; - when X4 and X' are CH2 and 'W' is a double bond, R7 is not CH20Ac, C02Me or H; (ii) when X1 is CHOH or CHOAc, X4 and X5 are CH2, "b" is a single bond, R1-5 are Me, - R7 is not COOH when R is CH2, R10 is CHO; and - R7 is not CH2OAc or C02Me, when R?, R10 are Me; when X' is C=O, X4and X' are CH2, "b" is a single bond, R'-5, R9 and R10 are Me, R7 is not COOH, Me or CH20Ac; (iv) when X' is CHOH, X4 and X are CH2, 'W' is a double bond, R'-', R, W' are Me, R is not COOH.
18. A compound according to claim 17 wherein R', R3-5 are methyl, R is H or methyl, and R' is defined as below for the relevant group R"; when X' is:
-CHOR la 5 R la is H; -CHCOY-Hal, Y is CH2 and Hal is Cl; when X5 is CHOCOR1b, R Ib is methyl; when R7 is: -CHOOR1c, W' is H or methyl; CO0Y0COR1c, Y is CH2 and W' is H; -CH2OR1c, R1c is H; -CH2OCOR1r, W' is methyl; When R9 is =CHRId CH 2 ORld or OR Id R Id is H, and when R9 is R 1d y R ld is methyl; when R10 is R", R le is methyl, and when R10 is COOR", COW' or CH2OW', R le is H.
19. A compound according to claims 17 or 18 selected from the table of claim 3.
20. A process for preparing a compound of formula 1, as defined in claim 1, wherein X4 is C=0 and X5 is C=0 or CH2, 0 H 1 F3 R4 00OR10 X H R, YH F R, comprising (i) oxidising a compound of formula Ib to form a compound of formula lc; and 1b 0 H 1 RRCOOF0.
R4 H 1 ( X,. H PR, H R, R, IC 0 H 1 0 cc R, R, -D(DF' 0 H CC go- X, t H #R, R2 R, H 1,4 Id 36 (ii) where W' is H, esterifying with an appropriate group bearing the desired W' substituent; and optionally oxidising the product of step (i) or (ii) to form a compound of formula Id.
21. A process according to claim 20 wherein the compound of formula Ib is oxidised to lc by treating with ruthenium tetroxide.
22. A process according to claims 20 or 21 wherein the compound of formula Ic is oxidised to Id by treating with selenium dioxide.
23. A processs for preparing a compound of formula 1 as defined in claim 1, wherein X4 and X5 are CH2, H R3 R 4 ICOOFc R H R5 X, - - H R 2 R, H-,_,,0 H R3 R4 COOF' X,. H R5 t H R2 R, 900 le If comprising oxidising a compound of formula le to form a compound of formula If
24. A process according to claim 23 wherein the compound of formula Ie is oxidised to If by treating with selenium dioxide.
GB0012823A 2000-05-23 2000-05-25 Triterpenoid derivatives Withdrawn GB2362649A (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
GB0012823A GB2362649A (en) 2000-05-25 2000-05-25 Triterpenoid derivatives
PCT/GB2001/002309 WO2001090046A1 (en) 2000-05-23 2001-05-23 Triterpenoid derivatives
EP01936618A EP1292562A1 (en) 2000-05-23 2001-05-23 Triterpenoid derivatives
AU2001262489A AU2001262489A1 (en) 2000-05-23 2001-05-23 Triterpenoid derivatives

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB0012823A GB2362649A (en) 2000-05-25 2000-05-25 Triterpenoid derivatives

Publications (2)

Publication Number Publication Date
GB0012823D0 GB0012823D0 (en) 2000-07-19
GB2362649A true GB2362649A (en) 2001-11-28

Family

ID=9892423

Family Applications (1)

Application Number Title Priority Date Filing Date
GB0012823A Withdrawn GB2362649A (en) 2000-05-23 2000-05-25 Triterpenoid derivatives

Country Status (1)

Country Link
GB (1) GB2362649A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996029068A2 (en) * 1995-03-21 1996-09-26 Board Of Trustees Of The University Of Illinois Use of betulinic acid and its derivatives for inhibiting malignant melanoma growth and a method of monitoring this
JPH1017431A (en) * 1996-07-02 1998-01-20 Pola Chem Ind Inc Skin ununiformity improver and cosmetic containing the same
WO1998032443A1 (en) * 1997-01-24 1998-07-30 Marigen S.A. Ultramicro-emulsions of spontaneously dispersible concentrates containing antitumorally, antivirally and antiparasitically active esters of pentacyclic triterpenes
WO2000024762A1 (en) * 1998-10-28 2000-05-04 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Betulinic acid and derivatives thereof useful for the treatment of neuroectodermal tumors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996029068A2 (en) * 1995-03-21 1996-09-26 Board Of Trustees Of The University Of Illinois Use of betulinic acid and its derivatives for inhibiting malignant melanoma growth and a method of monitoring this
JPH1017431A (en) * 1996-07-02 1998-01-20 Pola Chem Ind Inc Skin ununiformity improver and cosmetic containing the same
WO1998032443A1 (en) * 1997-01-24 1998-07-30 Marigen S.A. Ultramicro-emulsions of spontaneously dispersible concentrates containing antitumorally, antivirally and antiparasitically active esters of pentacyclic triterpenes
WO2000024762A1 (en) * 1998-10-28 2000-05-04 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Betulinic acid and derivatives thereof useful for the treatment of neuroectodermal tumors

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Chemical Abstracts 111:134551 *
Chemical Abstracts 115:138458 *
Chemical Abstracts 123:169934 *
Chemical Abstracts 128:140866 *
Chemical Abstracts 128:145138 and & JP 10 017431 A *

Also Published As

Publication number Publication date
GB0012823D0 (en) 2000-07-19

Similar Documents

Publication Publication Date Title
US7749988B2 (en) Triterpenoid derivatives
CN101117348B (en) A and C macrocyclic oxidation substituted pentacyclic triterpanoids and preparation method and use thereof
Borkova et al. Synthesis of cytotoxic 2, 2-difluoroderivatives of dihydrobetulinic acid and allobetulin and study of their impact on cancer cells
CN113234116B (en) Tripterine derivative, preparation method and medical application thereof
Wang et al. Studies on chemical structure modification and biology of a natural product, Gambogic acid (I): Synthesis and biological evaluation of oxidized analogues of gambogic acid
Guo et al. Synthesis, characterization and biological evaluation of some 16E-arylidene androstane derivatives as potential anticancer agents
Xu et al. Structure–activity relationship of Garcinia xanthones analogues: Potent Hsp90 inhibitors with cytotoxicity and antiangiogenesis activity
Selka et al. Discovery of a novel 2, 5-dihydroxycinnamic acid-based 5-lipoxygenase inhibitor that induces apoptosis and may impair autophagic flux in RCC4 renal cancer cells
CA3169315A1 (en) Glucose triptolide conjugates and uses thereof
Liu et al. Incorporation of nitric oxide donor into 1, 3-dioxyxanthones leads to synergistic anticancer activity
Li et al. Synthesis and discovery of mitochondria-targeting oleanolic acid derivatives for potential PI3K inhibition
WO2001090046A1 (en) Triterpenoid derivatives
Wang et al. Garcinol and its analogues: Synthesis, cytotoxic activity and mechanistic investigation
Zhan et al. Design and synthesis of a gossypol derivative with improved antitumor activities
Rárová et al. Effect of modification of betulinic acid at the C3-carbon atom of homolupane triterpenoids on the antiproliferative activity in vitro
Laube et al. Total synthesis of two 12-nordrimanes and the pharmacological active sesquiterpene hydroquinone yahazunol
Kádár et al. Efficient synthesis of novel A-ring-substituted 1, 2, 3-triazolylcholestane derivatives via catalytic azide-alkyne cycloaddition
US7858606B2 (en) Triterpenoid derivatives
GB2362649A (en) Triterpenoid derivatives
EP1289977B1 (en) Triterpenoid derivatives and their use as antiproliferative agents
Ni et al. Spirolactone-type and enmein-type derivatives as potential anti-cancer agents derived from oridonin
Hou et al. Synthesis of oridonin derivatives via mizoroki-heck reaction and click chemistry for cytotoxic activity
Wang et al. 5-(4-Hydroxyphenyl)-3H-1, 2-dithiole-3-thione derivatives of brefeldin A: Design, synthesis and cytotoxicity in MDA-MB-231 human breast cancer cells
Wang et al. Synthesis and preliminary anti-inflammatory activity exploration of novel derivatives of kirenol
Levina et al. Polar steroids from Solaster endeca starfish and the physiological activity of polar steroids from three starfish species

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)