WO2000004023A9 - Anhydride modified cantharidin analogues useful in the treatment of cancer - Google Patents

Anhydride modified cantharidin analogues useful in the treatment of cancer

Info

Publication number
WO2000004023A9
WO2000004023A9 PCT/AU1999/000567 AU9900567W WO0004023A9 WO 2000004023 A9 WO2000004023 A9 WO 2000004023A9 AU 9900567 W AU9900567 W AU 9900567W WO 0004023 A9 WO0004023 A9 WO 0004023A9
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cells
rule
cantharidin
treatment
Prior art date
Application number
PCT/AU1999/000567
Other languages
French (fr)
Other versions
WO2000004023A1 (en
Inventor
Adam Mccluskey
Jennette A Sakoff
Stephen Ackland
Alistair T R Sim
Original Assignee
Univ Newcastle Res Ass
Adam Mccluskey
Jennette A Sakoff
Stephen Ackland
Alistair T R Sim
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 Univ Newcastle Res Ass, Adam Mccluskey, Jennette A Sakoff, Stephen Ackland, Alistair T R Sim filed Critical Univ Newcastle Res Ass
Priority to JP2000560129A priority Critical patent/JP2002520415A/en
Priority to KR1020017000638A priority patent/KR20010074721A/en
Priority to AU47634/99A priority patent/AU772501B2/en
Priority to EP99930941A priority patent/EP1104423A4/en
Priority to CA002337771A priority patent/CA2337771A1/en
Publication of WO2000004023A1 publication Critical patent/WO2000004023A1/en
Publication of WO2000004023A9 publication Critical patent/WO2000004023A9/en
Priority to AU2004203476A priority patent/AU2004203476A1/en
Priority to US11/240,971 priority patent/US20060030616A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This invention relates to compounds useful in the treatment of certain forms of
  • the compounds are specific inhibitors of protein phosphatases 1 and
  • apoptosis cellular signal transduction, as neurotransmission, muscle contraction,
  • cdkl a universal protein kinase
  • checkpoints are responsible for halting cell cycle progression in G j S and/or G 2 phases
  • activation of cdk/cyclin complexes requires the phosphorylation of a
  • Cdc25 is only active in its
  • protein phosphatase 2 A (PP2A) can inhibit the
  • the G,/S checkpoint is predominantly regulated by the cdk/cyclin D/E complex
  • pRb protein retinoblastoma
  • E2F controls the transcription of proteins
  • protein phosphatase 1 can dephosphorylate pRb and
  • Serine/threonine phosphatases which are responsible for protein
  • dephosphorylation comprise a unique class of enzymes consisting of four primary
  • phosphatase 1 (PPl) and 2A (PP2A) inhibitors phosphatase 1 (PPl) and 2A (PP2A) inhibitors.
  • Okadaic acid is more specific for PP2A
  • the cell does not have the capacity to detect DNA damage or malformation of the
  • tumour promoter in some cell types, it has been shown to revert the tumour
  • okadaic acid has been shown to selectively enhance the cytotoxicity
  • calyculin enhances irradiation killing in fibroblast cells
  • okadaic acid has been shown to significantly increase the fraction of
  • dTMP via TS inhibition
  • okadaic acids class of compounds, with the exceptions of okadaic acid,
  • inside cells is such that high concentrations of these inhibitors are required to generate a
  • tumour suppressor The most commonly mutated gene in human cancers is the tumour suppressor
  • mutant p53 is certainly desirable, for two main reasons. Firstly, cells that have an
  • tumours such as colon carcinoma and non small cells
  • mutant p53 phenotype would potentially produce fewer side effects since only the
  • cancer cells would be killed and not the p53 proficient normal healthy cells.
  • R alkyl or aryl
  • SUBSTITUTE SHEET (RULE 26) being potent, selective, oxidatively stable, and cell permeable inhibitors of protein
  • permeable inhibitors of protein phosphatases 1 and 2A said inhibitors being anhydride
  • R, and R 2 are H, aryl or alkyl; X is O, N or S; Y is O, S, SR, NH, NR, CH 2 OH,
  • R 2 can cyclise to form a ring as follows:
  • R 3 and R 4 are H, aryl or alkyl.
  • the aryl group may suitably be phenyl or naphthyl for example, and may be
  • the alkyl group may be attached via a carbon spacer of between 6 and 10 carbon atoms.
  • the alkyl group may be
  • the process may include the
  • the process may further involve hydrogenation of the adduct of the diene and
  • cantharidin analogues are dependent on the aromaticity of the starting diene. Suitable
  • treating a cancer which method comprises administering to a patient in need of such
  • the method may be carried out in conjunction with one or more further steps
  • sensitising cancer cells to at least one method of treating cancer which method of
  • sensitising comprises administering to a patient in need of such treatment, an effective
  • treating cancer which method comprises:
  • anhydride modified cantharidin analogue to sensitise cancer cells of the patient to one or
  • the method may also comprise
  • the one or more cancer treatments mentioned above may be selected from
  • Figure 1 is a schematic representation of the structure activity data generated for
  • FIG. 1 New cantharidin analogues.
  • Figure 3 Cytotoxicity of cantharidin and the new cantharidin analogues.
  • Figure 5 Cell cycle analysis 18h after 6Gy of radiation and 12h after exposure to cantharidin, MK-2 or MK-4.
  • cantharidin analogues encompassed by the present invention generally depend on the
  • the solvent can be for example, ether (for room temperature
  • SUBSTITUTE SHEET (RULE 26) recrystalisation from for example, chloroform or ethanol.
  • SUBSTITUTE SHEET (RULE 26) mobile phase.
  • the complexes are precipitated by the addition of a saturated sodium tetraphenylborate solution.
  • tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non-invasive tumour types such as, but not limited to, colon cancer and non
  • the compounds of this invention are suitably administered intravenously,
  • diluents may be used in conjunction with the
  • Suitable such pharmaceutically acceptable substances are those within the
  • the dosage level can be readily determined by the physician in accordance with
  • the compounds of this invention may also sensitise cancer cells to other
  • these methods include irradiation and
  • platinum anti-cancer agents for example cisplatin.
  • sensitisation may also be brought about by, for example the use of
  • fluorouracil methotrexate and antifolates.
  • the compounds of this invention sensitise those cells with deficient
  • cancer cell lines may be chosen. These are typically both haematopoietic and
  • solid tumour cell lines with varying p53 status include: L1210 (murine leukaemia,
  • HL60 human leukaemia, p53 nul
  • A2780 human ovarian carcinoma
  • HCT116 human colon carcinoma, p53
  • the cancer cell cycle the following are suitably used:
  • the cells are fixed in 70% ethanol and stored at - 20°C until analysis is
  • fluorescence is collected in fluorescence detector 2 (FL2), filter 575/30 nm
  • yl]2,5-diphenyl-tetrazolium bromide assay is used to determine whether a synergistic
  • SUBSTITUTE SHEET (RULE 26) lines are plated at a density of 2000-25000 cells/well. The cells are left for 24h prior to
  • inhibitory effects are evaluated using the MTT assay and absorbance read at 540 nm.
  • the IC 50 is the drug concentration at which cell growth is 50% inhibited based on the
  • IC 5 ⁇ values the drug concentration at which cell growth is 50% inhibited.
  • P53 mutant cells are generally more resistant to conventional
  • the cells will be unable to initiate repair mechanisms and will be more unstable and
  • Cisplatin is another commonly used anticancer treatment which binds to DNA
  • Cisplatin is particularly useful in the
  • testicular carcinoma small cell carcinoma of the lung, bladder cancer, and
  • mutant cells is desirable since 50% of tumours have either a mutated or deleted p53
  • Thymidylate synthase TS
  • TS catalyses a critical
  • TS is a key regulatory enzyme during the S-phase of the cell cycle.
  • TS inhibitors such as fluorouracil, raltitrexed, and
  • LY231514 play a pivotal role in anticancer treatment and are often the first line
  • Thymitaq (Zarix, Ltd) be used in combination with cantharidin analogues. Thymitaq is a
  • Anhydride modified cantharidin analogues were synthesised by a variety of
  • cantharidin analogues have been synthesised.
  • the present inventors have
  • maleimide occupies the hydrogen bond cavity normally reserved for the 7-0 substituent
  • Z hydrophobic tail; eg long chain nitrile: cf Calyculin A, long chain terminating
  • membrane permeable protein phosphatase inhibitors are screened for their anti-cancer
  • Protein phosphatase 1 and 2A were partially purified from chicken skeletal
  • the total assay volume was 30 ml.
  • the assay conditions were restricted to
  • haematopoietic and solid tumour cell lines with varying p53 status include:
  • HL60 human leukaemia, p53 nul
  • A2780 human ovarian carcinoma, p53 wildtype
  • ADDP cisplatin resistant A2780 cells, p53 mutant
  • SW480 human colon carcinoma, p53 mutant
  • WiDr human colon carcinoma, p53 mutant
  • HT29 human colon carcinoma, p53 mutant
  • HCT1 16 human colon carcinoma, p53 wildtype
  • Anti-cancer screening of the protein phosphatase inhibitors is assessed using the
  • MTT assay determines cell viability by the ability of mitochondrial
  • MK-7 IC 50 247 to >1000 ⁇ M
  • Tumour Cell p53 IC-50 (mean + SE) after 72h continuous exposure ( ⁇ M) type line status Cantharidin MK-1 MK-2 MK-3 MK-4 MK-5 MK-7 MK-8 MK-9
  • MK-4 and MK-5 were selectively more cytotoxic in the human colon cancer
  • ovarian IC 50 275-333 ⁇ M; 260-567 ⁇ M
  • osteosarcoma IC 50 450 ⁇ M
  • checkpoint of the cell cycle may be determined by cell cycle analysis using flow
  • asynchronous cell cultures are harvested 18h after 6Gy irradiation
  • cantharidin is greater than for its analogues. Interestingly, cantharidin also showed slight
  • HL60, HT29 and HCTl 16 cells The cells were treated with 6Gy of radiation and then
  • MK-4 also abrogated G, arrest in L1210, HL60 and
  • HCTl 16 cells In HT29 cells, MK-4 induced abrogation of the G 2 checkpoint. It is
  • HCTl 16 and HT29 cells was additive and not synergistic using drug molar ratios of 1 : 1 ,
  • MK-4 in HT29 cells was also additive using drug molar ratios of 1 : 10, 1 : 100, 1 :1000
  • a synthetic inhibitor such as 3 represents a significant advance on the currently
  • dicarboxylic acid moiety gives rise to a PPl selective compound.
  • the retinoblastoma protein associates with the protein
  • the retinoblastoma protein a master regulator of

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Furan Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

Anhydride modified cantharidin analogues useful in the treatment of certain forms of cancer also methods for the screening for anti-cancer activity of these analogues and/or their ability to sensitise cancer cells to cancer treatment. The modified cantharidin analogues have structure (I) or (II), wherein R1, R2, R3 and R4 are H, aryl or alkyl; X is O, N or S; Y is O, S, NH, NR; R is alkyl or aryl; A and B are H or CH3; W and Z are CHOH or C=O. These compounds inhibit protein phosphatase.

Description

ANHYDRIDE MODIFIED CANTHARIDIN ANALOGUES USEFUL IN THE TREATMENT OF CANCER
TECHNICAL FIELD
This invention relates to compounds useful in the treatment of certain forms of
cancer; processes for producing these compounds; methods of treatment using these
compounds per se; methods of treatment using these compounds which methods also
increase the sensitivity of cancer cells to other treatments; methods of screening these
compounds for anti-cancer activity; and methods of screening these compounds for
anti-cancer activity and/or ability to sensitise cancer cells to other methods of treatment.
More particularly, the compounds are specific inhibitors of protein phosphatases 1 and
2 A.
BACKGROUND ART
Protein phosphatase inhibitors and the abrogation of cell cycle checkpoints
The regulation of protein phosphatases is integral to the control of many cell
processes, including cell growth, transformation, tumour suppression, gene transcription,
apoptosis. cellular signal transduction, as neurotransmission, muscle contraction,
glycogen synthesis, and T-cell activation. The role of protein phosphatases in many of
these processes is often mediated via alterations in the cell cycle. Cell cycle progression
is tightly regulated to ensure the integrity of the genome. During cell division it is
imperative that each stage of the cell cycle be completed before entry into the next, and
this is achieved tluough a series of checkpoints. The cell cycle can be broken down into
four phases, the first gap (G^, is followed by a phase of DNA synthesis (S-phase); this is
followed by a second gap (G ) which in turn is followed by mitosis (M) which produces
two daughter cells in G). There are two major control points in the cell cycle, one late in
SUBSTITUTE SHEET (RULE 26) G and the other at the G2/M boundary. Passage through these control points is
controlled by a universal protein kinase, cdkl . The kinase activity of cdkl is dependant
on phosphorylation and the association with a regulatory subunit, cyclin B. The periodic
association of different cyclins with different cyclin dependent kinases (cdk) has been
shown to drive different phases of the cell cycle; thus cdk4-cyclin Dl drives cells
through mid Gb cdk2-cyclin E drives cells in late Gb cdk2-cyclin A controls entry into
S-phase and cdc2-cyclin B drives the G2/M transition (O'Connor, 1996, 1997).
Following DNA damage induced by chemotherapy or radiation treatment these
checkpoints are responsible for halting cell cycle progression in Gj S and/or G2 phases
(O'Connor, 1996). The cell undergoes a cell cycle arrest so that the damaged DNA can
be repaired before entry into S phase or mitosis. The phase at which the cell cycle is
halted will depend upon the type of DNA damaging agent used and the point during the
cell cycle that the damage was incurred (O'Connor, 1997). The cell cycle is controlled
and regulated by an intricate phosphorylation network (Stein et al., 1998). More
particularly, activation of cdk/cyclin complexes requires the phosphorylation of a
conserved threonine residue, which are catalysed by CAK kinase, as well as the removal
of inhibitory phosphorylations by the phosphatase cdc25. Cdc25 is only active in its
phosphorylated form. Therefore, protein phosphatase 2 A (PP2A) can inhibit the
activation of cdk/cyclin complexes by inhibiting CAK activity and by dephosphorylating
cdc25. The G,/S checkpoint is predominantly regulated by the cdk/cyclin D/E complex
that mediates its effects by phosphorylating and inactivating the tumour suppressor
protein retinoblastoma (pRb). The phosphorylation of pRb prevents it from interacting
with the S-phase transcription factor E2F. E2F controls the transcription of proteins
SUBSTITUTE SHEET (RULE 26) - j - needed for DNA synthesis and entry into S-phase including thymidylate synthase.
Accordingly, the inactivation of pRb by phosphorylation permits entry into the S-phase
and vice versa. However, protein phosphatase 1 (PPl) can dephosphorylate pRb and
inhibit the cell cycle (Durfee et al., 1993). Thus, PPl and PP2A are both negative
regulators of the cell cycle. Inhibition of PPl and PP2A would abrogate these
checkpoints and prematurely force cells through the cell cycle.
Serine/threonine phosphatases, which are responsible for protein
dephosphorylation. comprise a unique class of enzymes consisting of four primary
subclasses based on their differences in substrate specificity and environmental
requirements. Of the serine/threonine phosphatases, protein phosphatases 1 and 2 A (PPl
and PP2A, respectively) share sequence identity between both enzyme subunits (50% for
residues 23-292; 43% overall), are present in all eukaryotic cells and are together
responsible for 90% of all cellular dephosphorylation. Knowledge of structure and
subsequent correlation of binding function for both PP 1 and PP2 A would therefore
provide a vital link toward understanding the biochemical role of these enzymes. A goal
of the medicinal chemist is the development of potent and selective inhibitors of these
protein phosphatases.
The natural toxins, okadaic acid, calyculin A, microcystin-LR and tautomycin are
representative of a structurally diverse group of compounds that are all potent protein
phosphatase 1 (PPl) and 2A (PP2A) inhibitors. Okadaic acid is more specific for PP2A
(IC50 InM) than PPl (IC50 60nM), while calyculin is slightly more specific for PPl (IC50
0.5-1. OnM) than PP2A (IC50 2nM). All of these phosphatase inhibitors are known to
abrogate cell cycle checkpoints, particularly the G2 checkpoint of the cell cycle and
SUBSTITUTE SHEET (RULE 26) induce cellular mitoses (Yamashita et al., 1990). Abrogation of the G2 checkpoint means
that the cell does not have the capacity to detect DNA damage or malformation of the
genome prior to entry into mitosis. Therefore, cells which have a deficient G2 checkpoint
are unstable, and incapable of detecting DNA damage, initiating G2 arrest, or undergoing
DNA repair. Such cells enter the mitotic stage of the cell cycle prematurely with
malformed spindles. The abrogation is of the G2 checkpoint in the cell cycle by okadaic
acid is mediated via the activation of cdc2/Hl kinase. the major mitotic inducer, and
results in a premature mitotic state (Yamashita et al., 1990). Although okadaic acid is
known as a tumour promoter, in some cell types, it has been shown to revert the
phenotype of oncogene-transformed cells to that of normal cells, and to inhibit neoplastic
transformation of fibroblasts (Schonthal, 1991 ).
Furthermore, okadaic acid has been shown to selectively enhance the cytotoxicity
of vinblastine and the formation of apoptotic cells, in HL60 cells which are p53 nul
(Kawamura, 1996). Interestingly, calyculin enhances irradiation killing in fibroblast cells
at doses that are non toxic when given as a single treatment. (Nakamura and Antoku,
1994). Data also shows that okadaic acid can abrogate the G,/S checkpoint of the cell
cycle. In this context, okadaic acid has been shown to overide the S-phase checkpoint
and accelerate progression of G2-phase to induce premature mitosis (Gosh et al., 1996).
In addition, okadaic acid has been shown to significantly increase the fraction of
quiescent cells entering the S-phase via modifications in the phosphorylation state of
pRb (Lazzereschi et al., 1997). Other studies have shown that the hyperphosphoryation
state of pRb forces cells prematurely into S-phase and pRb can be kept in a
phosphorylated state via protein phosphate inhibition (Herwig and Strauss, 1997). Cells
SUBSTITUTE SHEET (RULE 26) lacking functional pRb show increased apoptosis and cytotoxicity following 5-
fluorouracil and methotrexate treatment (Herwig and Strauss, 1997). We propose that
cell death would be substantially enhanced in cells forced to enter the S-phase
prematurely (via G, checkpoint abrogation) and which were lacking key S-phase
components such as dTMP (via TS inhibition).
The okadaic acids class of compounds, with the exceptions of okadaic acid,
cantharidin (Honaken) and thyrisferyl 23 -acetate (Matszawa et. al) (being PP2A
selective) exhibit poor selectivity. Furthermore, the concentration of PPl and PP2A
inside cells is such that high concentrations of these inhibitors are required to generate a
response in vivo resulting in the loss of effectiveness of any in vitro selectivity (Wang).
Cantharidin (exo.exo-2.3-dimethyl-7-oxobicyclo[2.2.1]heptane-2,3-dicarboxylic
acid anhydride), is a major component of the Chinese blister beetles:
Mylabris phalerata or M. cichorii)(Yang; Cavill et. al). The dried body of these beetles
has been used by the Chinese as a natural remedy for the past 2000 years. Although
Western medicine decreed cantharidin to be too toxic in the early 1900's (Goldfarb et.
al) its purported aphrodisiac qualities (the active ingredient of "Spanish Fly"), and its
widespread occurrence in cattle feed still results in numerous human and livestock
poisonings (Schmitz).
Li and Casida, and previous work in this laboratory (McCluskey et. al) (and
more recently Pombo-Villar, Sodeoka) has assisted in the delineation of certain features
crucial for inhibition of PP2A by cantharidin analogues (Figure 1). However the
corresponding picture for PPl is not so clear, the majority of data refers to possible
SUBSTITUTE SHEET (RULE 26) interactions with the known crystal structures, and in some cases the inhibition values
for PPl are not reported.
Involvement of Tumour Suppressor Gene p53
The most commonly mutated gene in human cancers is the tumour suppressor
gene p53, which is abnormally expressed in more than 50% of tumours. The
development of chemotherapeutic agents which selectively target cancer cells with
mutant p53 is certainly desirable, for two main reasons. Firstly, cells that have an
abnormal p53 status are inherently resistant to conventional chemotherapy and produce
the more common, and more aggressive tumours such as colon carcinoma and non small
cell lung cancer. Secondly, a chemotherapy regime that targeted only those cells with a
mutant p53 phenotype would potentially produce fewer side effects since only the
cancer cells would be killed and not the p53 proficient normal healthy cells.
DISCLOSURE OF THE INVENTION
In relation to the discussion above, the present inventors believed that the
replacement of the ether O atom of the anhydride with N or S (as N-H and N-R, where
R = alkyl or aryl) would allow them to probe the H-bonding requirements of this region
of cantharidin analogues. Previous studies in their laboratory had shown limited
tolerance for modification of the 7-oxa position. An ability to modify these heteroatoms
is crucial to the development of selective inhibitors based on this simple skeleton.
There is not, at present, an inhibitor with either absolute specificity or high
enough selectivity which renders the inhibitor effectively specific in vivo.
It has surprisingly been found that anhydride modified cantharidin analogues,
which are the subject of this invention, may possess one or more of the properties of
SUBSTITUTE SHEET (RULE 26) being potent, selective, oxidatively stable, and cell permeable inhibitors of protein
phosphatases 1 and 2A.
Therefore, according to the first aspect of this invention there are provided cell
permeable inhibitors of protein phosphatases 1 and 2A, said inhibitors being anhydride
modified cantharidin analogues.
According to a particular embodiment of the first aspect of this invention there
are provided compounds of the formula:
Figure imgf000009_0001
wherein R, and R2 are H, aryl or alkyl; X is O, N or S; Y is O, S, SR, NH, NR, CH2OH,
CH2OR; R is alkyl or aryl; A and B are H or CH3; W and Z are CHOH or C=0 and R
and R2 can cyclise to form a ring as follows:
Figure imgf000009_0002
wherein R3 and R4 are H, aryl or alkyl.
The aryl group may suitably be phenyl or naphthyl for example, and may be
attached via a carbon spacer of between 6 and 10 carbon atoms. The alkyl group may
suitably be C C^.
SUBSTITUTE SHEET (RULE 26) According to the second aspect of this invention there is provided a process for
producing anhydride modified cantharidin analogues. The process may include the
steps of:
dissolving a diene in a suitable solvent and adding to the resultant solution an
ene.
According to a third aspect of the invention there is provided a process for
producing anhydride modified cantharidin analogues, involving the step of reacting a
diene with an ene.
The process may further involve hydrogenation of the adduct of the diene and
ene and/or optionally, ring opening of the adduct.
Generally, the reaction conditions for the production of the anhydride modified
cantharidin analogues are dependent on the aromaticity of the starting diene. Suitable
reaction conditions are exemplified below.
According to a fourth aspect of this invention there is provided a method of
treating a cancer which method comprises administering to a patient in need of such
treatment, an effective amount of an anhydride modified cantharidin analogue of the
first aspect of this invention, together with a pharmaceutically acceptable carrier, diluent
and/or excipient.
The method may be carried out in conjunction with one or more further
treatments for treating the cancer.
According to a fifth aspect of this invention there is provided a method of
sensitising cancer cells to at least one method of treating cancer, which method of
sensitising comprises administering to a patient in need of such treatment, an effective
SUBSTITUTE SHEET (RULE 26) amount of an anhydride modified cantharidin analogue of the first aspect of this
invention, together with a pharmaceutically acceptable carrier, diluent and/or excipient.
According to a sixth aspect of the invention there is provided a method of
treating cancer which method comprises:
administering to a patient in need of such treatment, an effective amount of an
anhydride modified cantharidin analogue to sensitise cancer cells of the patient to one or
more cancer treatments; and utilising the one or more cancer treatments.
According to a seventh aspect of this invention there is provided a method of
screening a compound for anti-cancer activity.
According to an eighth aspect of this invention there is provided a method of
screening compounds for use in the fourth aspect of this invention, said method
comprising screening for anti-cancer activity; and screening for ability to abrogate either
the G| or the G2 checkpoint of the cancer cell cycle. The method may also comprise
screening for the ability of said compounds to sensitise cancer cells to one or more
cancer treatments.
The one or more cancer treatments mentioned above may be selected from
treatments involving cisplatin, irradiation, taxanes and antimetabolites.
The invention will hereinafter be described with reference to Examples and the
accompanying figures.
Brief Description of the Figures
Figure 1 is a schematic representation of the structure activity data generated for
inhibition by PP2A by cantharidin analogues;
Figure 2: New cantharidin analogues.
Figure 3: Cytotoxicity of cantharidin and the new cantharidin analogues.
SUBSTITUTE SHEET (RULE 26) Figure 4: Cell cycle analysis 12h following exposure to cantharidin, MK-2 or MK-4.
Figure 5: Cell cycle analysis 18h after 6Gy of radiation and 12h after exposure to cantharidin, MK-2 or MK-4. Figure 6 (a-c): Combination index versus fraction affected: HCT116 colon cells in simultaneous combination with cisplatin and MK-4.
Figure 7 (a-b): Combination index versus fraction affected: HT29 colon cells in simultaneous combination with cisplatin and MK-4.
Figure 8 (a-c): Combination index versus fraction affected: HCT116 colon cells in simultaneous combination with taxotere and MK-4
Figure 9 (a-c): Combination index versus fraction affected: HT29 colon cells in
simultaneous combination with taxotere and MK-4.
Best and other Modes for Carrying Out the Invention
As mentioned above, the reaction conditions for producing anhydride modified
cantharidin analogues encompassed by the present invention generally depend on the
aromaticity of the starting diene. This is illustrated by a description of examples of the
methods wherein the starting materials are furan (Method 1 below): thiophene (Method
2 below); and pyrrole (Method 3 below).
Method 1 : Furan as the starting diene
A solution of furan (5 equivalents) is dissolved in a suitable solvent (about 5 times
the volume of furan, the solvent can be for example, ether (for room temperature
reactions); or benzene or xylene (the latter two for reactions at 80 and 130°C
respectively). To this solution is added one equivalent of the ene. The reaction is then
heated (or stirred at room temperature), typically for 24 hours (2 days in the case of the
room temperature reaction). Upon cooling (or standing at room temperature) a precipitate
forms and is collected by vacuum filtration. The adduct is then purified by
SUBSTITUTE SHEET (RULE 26) recrystalisation from for example, chloroform or ethanol. In the case of the furan +
maleic anhydride compound care is exercised to minimise heating as this causes a reto-
Diels-Alder reaction yielding only the starting materials.
Method 2: Thiophene as the starting diene
Thiophene (l .Olόg, 0.012 mol) and maleic anhydride (0.558.0.006 mol) are
mixed at room temperature in 2.5 mL of distilled dichloromethane. This mixture is then
placed inside a high pressure reactor. They are compressed to a pressure of 17kbar at
40°C for a period of 71 hours, after which the pressure is released and the product
purified by chromatography.
Method 3 : Pyrrole as the starting diene
To [Os (NH3)5OsO2 CF3 )] (CF3SO3)2 , (0.3511 g, 0.4 mmol) and activated
magnesium (0.151 1 g), pyrrole (0.45 mL, 0.6 mmol), DME (1 mL) and DMAc (0.3 mL)
are added in that order. The mixture is stirred for 1 hour, the temperature gradually
rising to 40°C and then dropping. The brown slurry is filtered through a thin pad of
celite, and the cake washed with DME in small portions (4 x 2 mL). The filtrate is
added to dichloromethane (15 mL). Vigorous stirring results in the formation of yellow
coloured precipitate which is collected by vacuum filtration, followed by an ether wash
(2 x 2.5 mL). The product is dried under a stream of nitrogen yielding a yellow-tan
solid (0.343g, 84%). To this pyrrole complex is added maleimide (0.05g, 0.515 mmol)
(or any other "ene", eg maleic anhydride, dimethyl maleate, etc) in acetonitrile. The
mixture is allowed to stir at room temperature for 60 min. after which the solvent is
removed by vacuum, yielding the exo isomer (0.359g, 64%). The crude material is
purified by ion-exchange column (Sephadex-CM C-25, 2 x 10 cm), using NaCl as the
SUBSTITUTE SHEET (RULE 26) mobile phase. The complexes are precipitated by the addition of a saturated sodium tetraphenylborate solution.
The types of cancer which are amenable to treatment by these compounds
include those types of cancer which are inherently resistant to conventional
chemotherapy. Typically, these types of cancer are represented by the more common
and more aggressive tumour types such as, but not limited to, colon cancer and non
small-cell lung cancer.
The compounds of this invention are suitably administered intravenously,
although other modes of administration are possible. Pharmaceutically acceptable
diluents, adjuvants, carriers and/or excipients may be used in conjunction with the
compounds of this invention.
Suitable such pharmaceutically acceptable substances are those within the
knowledge of the skilled person and include compounds, materials and compositions
deemed appropriate.
Actual dosage levels of the compounds of the invention may be varied so as to
obtain an amount of the active ingredient which is effective to achieve the desired
response for a particular patient, composition and mode of administration.
The dosage level can be readily determined by the physician in accordance with
conventional practices and will depend upon a variety of factors including the activity of
the particular compound of the invention to the administered, the route of
administration, the time of administration, the rate of excretion of the particular
compound employed, the age, sex, weight, condition, general health and prior medical
history of the patient being treated, and like factors well known in the medical arts.
SUBSTITUTE SHEET (RULE 26) The compounds of this invention may also sensitise cancer cells to other
methods of treatment. For example, typically these methods include irradiation and
treatment with platinum anti-cancer agents, for example cisplatin.
In addition, sensitisation may also be brought about by, for example the use of
the plant alkaloids vinblastine and vincristine, both of which interfere with tubulin and
the formation of the mitotic spindle, as well as taxanes and antimetabolites, including 5-
fluorouracil, methotrexate and antifolates.
In particular, the compounds of this invention sensitise those cells with deficient
p53 activity.
When screening for anti-cancer activity as contemplated by the invention,
various cancer cell lines may be chosen. These are typically both haematopoietic and
solid tumour cell lines with varying p53 status and include: L1210 (murine leukaemia,
p53 wildtype), HL60 (human leukaemia, p53 nul), A2780 (human ovarian carcinoma,
p53 wildtype), ADDP (cisplatin resistant A2780 cells, p53 mutant), SW480 (human
colon carcinoma, p53 mutant), WiDr (human colon carcinoma, p53 mutant), HT29
(human colon carcinoma, p53 mutant), HCT116 (human colon carcinoma, p53
wildtype) and 143B (human osteosarcoma, p53 mutant).
In addition to the methods for screening for anti-cancer activity, the following
procedures may be suitably used in the remainder of the screening process. For
example, when screening for the ability to abrogate the Gj and/or the G2 checkpoint of
the cancer cell cycle, the following are suitably used:
SUBSTITUTE SHEET (RULE 26) Cell cycle method
The cells are fixed in 70% ethanol and stored at - 20°C until analysis is
performed (1-2 weeks). After fixing, the cells are pelleted and incubated in PBS
containing propidium iodide (40mg/ml) and RNase A (200 mg/ml) for at least 30 min at
room temperature. The samples (2 X 10 events) are analysed using a Becton Dickson
FACScan, fluorescence is collected in fluorescence detector 2 (FL2), filter 575/30 nm
band pass. Cell cycle distribution is assessed using Cell Quest software (Becton
Dickson).
Those protein phosphatase inhibitors which show abrogation of either the Gj or
G2 checkpoint will then be exploited in combination studies with either radiation
exposure or chemotherapy drugs incubation. The MTT (3-[4,5-dimethylthiazol-2-
yl]2,5-diphenyl-tetrazolium bromide) assay is used to determine whether a synergistic,
antagonistic or additive effect is induced. The Median Effect method is adopted to
mathematically determine the optimal combination index of the treatments chosen
(Chou and Talalay, 1984). This method has been extensively used to investigate the
cytotoxicity of various drug combinations including cisplatin and D1694 (Ackland et al
1996; 1998). A combination index value less than 1 indicates synergism, a value equal
to 1 indicates additivity and a value greater than one indicates antagonism.
Cytotoxicity assay
When screening for the ability to sensitise cancer cells to conventional
chemotherapy and irradiation, the following methods are suitably used:
Cells in a subconfluent phase are transferred to 96- well microtitre plates. LI 210
cells are plated at a density of 1000 cells/well in lOOμl medium, while all other cell
SUBSTITUTE SHEET (RULE 26) lines are plated at a density of 2000-25000 cells/well. The cells are left for 24h prior to
treatment to ensure exponential growth has been achieved, 24h after plating (day 0),
lOOμl of phosphatase inhibitor is added to each well, control wells received lOOμl of
medium only. Drug exposure time is 72h (day 3). The effect of phosphatase inhibition
is tested in triplicate over a concentration range of 1 x 10"3M - 1 x 10"8 M. Growth
inhibitory effects are evaluated using the MTT assay and absorbance read at 540 nm.
The IC50 is the drug concentration at which cell growth is 50% inhibited based on the
difference of optical density on day 0 and day 3 of drug exposure. Cytotoxicity is
evaluated using a spectrophotometric assay which determines the percentage of cell
growth following exposure of the cells to various concentrations of the phosphatase
inhibitors for a period of 72 hours. The subsequent dose response curve is used to
calculate IC values (the drug concentration at which cell growth is 50% inhibited).
Most drug discovery has focused on the development of new single agents.
However, in light of the success of combination chemotherapy it is increasingly
apparent that successful anticancer treatment of the future will be based upon the
discovery of agents which are synergistic in their action. In view of this, the cytotoxicity
of phosphatase inhibitors in combination with either radiation, cisplatin, taxanes,
antimetabolites or plant alkaloids is examined. As indicated above, calyculin which by
itself is not cytotoxic, enhances irradiation induced cell death. Similarly abrogation of
the G2 checkpoint by either, caffeine or UCN-01, also enhances the cytotoxicity of γ
irradiation in cells with mutant p53 (CA46 and HT-29 cells) (Powell et al., 1995;
Russell et al.. 1995: Wang et al., 1996). DNA damage induced by irradiation causes
both a G, and G2 cell cycle arrest. In p53 mutant cells, the Gj checkpoint is absent.
SUBSTITUTE SHEET (RULE 26) However, following irradiation the cells will still arrest in the G2 phase, and potentially
repair the damage. P53 mutant cells are generally more resistant to conventional
chemotherapy and produce more aggressive tumours. Therefore, in p53 deficient cells,
DNA damage that is not detected by the G, checkpoint will be picked up by the G2
checkpoint. If the cells are deficient in both of these checkpoints then it is believed that
the cells will be unable to initiate repair mechanisms and will be more unstable and
increasingly susceptible to cell death induced by DNA damage.
Cisplatin is another commonly used anticancer treatment which binds to DNA
and produces DNA crosslinks and strand breaks. Cisplatin is particularly useful in the
treatment of testicular carcinoma, small cell carcinoma of the lung, bladder cancer, and
ovarian cancer. Repair of cisplatin induced DNA damage is mediated via nucleotide
excision repair which is coordinated by p53 activation of Gadd45 (Smith et al., 1994).
In this context, it has been suggested that cells that are p53 mutant are more sensitive to
cisplatin treatment (Hawkins et al., 1996). A number of researchers have investigated
this proposal in p53 mutant cell lines and in p53 mutant tumours, with mixed results.
While it is apparent that cisplatin is more cytotoxic in cells lines that are deficient in p53
(induced via papillomavirus) compared to the p53 proficient cells (Hawkins et al.,
1996), it is harder to test this hypothesis in tumours and in cisplatin resistant cells as
they may have several undefined mutations in their genome which would confound such
studies (Herod et al., 1996). Nevertheless, the G2 abrogator UCN-01 (7-
hydroxystaurosporine, a protein kinase inhibitor) has been shown to markedly enhanced
the cell-killing activity of cisplatin in MCF-7 cells defective for p53 function (Wang et
al., 1996).
SUBSTITUTE SHEET (RULE 26) The development of chemotherapeutic agents which selectively target p53
mutant cells is desirable since 50% of tumours have either a mutated or deleted p53
gene. Many of these p53 deficient cells and tumours are inherently resistant to
conventional chemotherapy and represent the common more aggressive tumour types
such as colon cancer, and non-small cell lung cancer. Thymidylate synthase (TS)
inhibitors are another class of commonly used anticancer agents. TS catalyses a critical
step in the pathway of DNA synthesis by converting dUMP to dTMP by methylation
using the co-substrate N5,N10-methylene tetrahydrofolate (CH2-THF) as a methyl
donor. This step is the only de novo source of dTMP, which is subsequently metabolised
to dTTP exclusively for incorporation into DNA during synthesis and repair (Jackman &
Calvert, 1995). Thus, TS is a key regulatory enzyme during the S-phase of the cell cycle.
Lack of dTTP results in DNA damage and ultimately cell death, but the process(es) by
which cell death occurs is not clear. TS inhibitors such as fluorouracil, raltitrexed, and
LY231514 play a pivotal role in anticancer treatment and are often the first line
treatment of many cancers (Peters & Ackland, 1996). We propose that the TS inhibitor
Thymitaq (Zarix, Ltd) be used in combination with cantharidin analogues. Thymitaq is a
direct and specific TS inhibitor which does not require active transport into the cell nor
does it require intracellular activation for its action.
The following examples are not to be construed as limiting on the scope of the
invention as indicated above.
SUBSTITUTE SHEET (RULE 26) Example 1
Chemistry
Anhydride modified cantharidin analogues were synthesised by a variety of
modified literature procedures, as set out in schemes 1 and 2. These modifications are
embodied in the three methods, which depend on the aromaticity of the starting dienes,
set out above. The dimethyl ester (3), which was prepared by the application of high
pressure, 17kbar, 40°C, 61 hours, as shown in scheme 3.
Figure imgf000020_0001
Scheme 1. a. Furammaleic anhydride (5:1). diethylether. 2d. RT, 96%; b. H2 / 10% Pd-
C/ EtOH; c. p-TosOH, MeOH, chromatography; d. H2 / 10% Pd-C/ Acetone; e. NaBH4
then HC1.
Figure imgf000020_0002
SUBSTITUTE SHEET (RULE 26) Scheme 2. Reagents and Conditions: f. Fura maleimide (5:1), diethyl ether, 7d, in
dark, 75%, exo product; g. FuramMaleimide (5: 1), diethylether, sealed tube 12h, 90°C,
66%,endo product.
Figure imgf000021_0001
Scheme 3. Reagents and Conditions: h.
Furan:dimethylmaleate (2:1), CH2C12 ,17 Kbar, 40°C. 61 h, 56%.
Example 2
Development of potent, selective, oxidativelv stable, and cell permeable inhibitors of
protein Phosphatases 1 and 2A.
Crude natural product extracts have yielded isopalinurin and a series of
cantharidin analogues have been synthesised. In this context, the present inventors have
developed the simple cantharidin analogue which is PPl selective (IC50 = 50mM, with
0% inhibition of PP2A at concentrations >1000mM) representing the first small
molecule to exhibit selectivity for PP 1. Results have indicated that a series of simple
synthetic modification of the cantharidin skeleton also allows the synthesis of a PP2A
selective compound (see Figure 1 ).
The present inventors have previously demonstrated that a facile ring opening of
an anhydride is crucial to inhibition of PP2 A. This is not possible with c (previous
studies with the 7-0, and this analogue indicated considerable hydrolytic stability of the
maleimide link). It is also interesting to note that endothal thioanhydride is three fold
more potent than cantharidin, with the S atom being an important factor. It is thus
SUBSTITUTE SHEET (RULE 26) envisaged that the 7-S group presents itself to the active sites metals and the N-H of the
maleimide occupies the hydrogen bond cavity normally reserved for the 7-0 substituent
cantharidin.
Structure of cantharidin and selective analogues
Figure imgf000022_0001
(a) (b)
(c)
(a) Shows structure of cantharidin;
(b) Shows PP 1 selective analogue; and
(c) Shows PP2A selective analogue. In the case of panel (c) IC50 ~ 25mM.
On the basis of these results and previous experience in our laboratory (synthesis
and molecular modelling of cantharidin inhibitors at PPl and PP2A), we have designed a
series of analogues which are more active and selective, whilst retaining the desirable
properties of stability and cell permeability.
The synthetic pathways to these analogues are shown in schemes 1-3. Each
scheme allows for modification of the basic skeleton, and in some cases the insertion of
beneficial feature that were present in the more complex natural toxin(s) (eg okadaic
acid, calyculin, microcystin, etc). The inclusion of these features is designed to provide
enhanced selectivity and potency.
SUBSTITUTE SHEET (RULE 26)
Figure imgf000023_0001
Example 3
Synthetic development of a series of PPl and PP2A analogues of cantharidin.
(i) Diels-Alder addition (maleic anhydride) and subsequent manipulations of X;
(ii) Diels-Alder addition (substituted maleic anhydrides), introduction and manipulation
of Z (Z = hydrophobic tail; eg long chain nitrile: cf Calyculin A, long chain terminating
in a spiro acetal: cf Tautomycm. Okadaic acid; long chain terminating in an aromatic
ring: cf Adda in Microcystin-LR; (iii) stereospecific ring opening of the anhydride
allowing further manipulations of the newly released functional groups (see scheme 2).
In this instance we have developed synthetic protocols in our laboratory that
allow the facile assembly of these analogues. Biological evaluation and molecular
modelling of the most active molecules will allow compounds to be evaluated.
Additional modification to the basic structure can be obtained as exemplified
below.
SUBSTITUTE SHEET (RULE 26)
Figure imgf000024_0001
X = CH2, S, O
Example 4
A specific example of one class of cantharidin analogue that shows promise as a
selective inhibitor of protein phosphatases 1 and 2A.
Figure imgf000024_0002
Example 5
Stereospecific route towards 7-azabicvclo [2.2.11 heptanes
We have shown that the introduction of the bridgehead nitrogen improves the
potency, selectivity and stability of similar analogues, the above pathway has been
developed to further improve the bio-activity of these analogues. The synthetic routes
alluded to herein may allow the rapid assembly of the target molecules.
Those agents which meet the requirements of being stable, specific, potent, and
membrane permeable protein phosphatase inhibitors are screened for their anti-cancer
activity.
SUBSTITUTE SHEET (RULE 26) Example 6
Biochemistry
All synthesised compounds were tested for their ability to inhibit protein
phosphatases 1 and 2A. Initial investigations were carried out at 100 mM. Promising
analogues were then assayed in triplicate for estimation of IC50 values.
Protein phosphatase 1 and 2A were partially purified from chicken skeletal
muscle essentially as described by Cohen Protein phosphatase activity was measured at
37°C in 50 mM Tris-HCl buffer (pH 7.4). 0.1 mM EDTA, 5 mM caffeine, 0.1% 2-
mercaptoethanol and 1 mg/ml bovine serum albumin using 30 mg [J P] -phosphorylase
as substrate. The total assay volume was 30 ml. The assay conditions were restricted to
20% dephosphorylation to ensure linearity and inhibition of protein phosphatase activity
was determined by including cantharidin or its analogues at the required concentrations
in the reaction buffer. Reactions were terminated by the addition of 0.1 ml ice cold 20%
trichloroacetic acid. Precipitated protein was pelleted by centrifugation and the
radioactivity in the supernatant measured by liquid scintillation counting. Data is
expressed as the percentage inhibition with respect to a control (absence of a competing
compound) incubation.
Example 7
Screening various PPl and PP2A inhibitors for anti-cancer activity
(a) Cytotoxicity of protein phosphatase inhibition:
Those PPl and PP2A inhibitors which fulfil the requirements detailed above
were tested in various cancer cell lines. The cells lines chosen for study included both
haematopoietic and solid tumour cell lines with varying p53 status and include:
SUBSTITUTE SHEET (RULE 26) L1210 (murine leukaemia, p53 wildtype),
HL60 (human leukaemia, p53 nul),
A2780 (human ovarian carcinoma, p53 wildtype),
ADDP (cisplatin resistant A2780 cells, p53 mutant),
SW480 (human colon carcinoma, p53 mutant),
WiDr (human colon carcinoma, p53 mutant).
HT29 (human colon carcinoma, p53 mutant)
HCT1 16 (human colon carcinoma, p53 wildtype)
143B (human osteosarcoma, p53 mutant)
Anti-cancer screening of the protein phosphatase inhibitors is assessed using the
MTT assay. This assay determines cell viability by the ability of mitochondrial
dehydrogenase to produce formazan crystals from 3-(4,5-dimethylthiazol-2-yl) -2, 5-
diphenyltetrazolium bromide. The viable cell number/well is directly proportional to the
production of formazan, which following solubilization, can be measured
spectrophotometrically (540nm). This technique is also used by the National Cancer
Institute to screen for new anticancer agents.
As described herein a number of cantharidin analogues have been synthesised
and tested for their anticancer activity in nine cancer cell lines using the MTT assay after
72 h exposure. These new analogues are shown in Figure 2 and have been designated
MK-1 through to MK-9. The cytotoxicity (IC50) of these cantharidin analogues is shown
in Table 1 and Figure 3. In summary, the MK-1 analogue did not show any significant
cytotoxicity in any of the cell lines tested (IC50 >1000μM). Only marginal cytotoxicity
across all cell lines tested was observed for MK-3 (IC50 247 to >1000μM), MK-7 (IC50
SUBSTITUTE SHEET (RULE 26) Table 1
ICso values of tumour cell lines after 72 h continuous exposure to cantharidin and cantharidin analogues.
Tumour Cell p53 IC-50 (mean + SE) after 72h continuous exposure (μM) type line status Cantharidin MK-1 MK-2 MK-3 MK-4 MK-5 MK-7 MK-8 MK-9
Murine Leukaemia L1210 wt 18 ± >1000 185 ±51 647 ± 132 680 ± 97 >1000 367 ± 37 337 ± 19 192 ±56
Human Leukaemia HL60 nul 13 ± >1000 177 ±3 247 ± 55 393 ± 103 323 ± 13 293 ±7 297 ±3 133 ±9
Human Ovarian A2780 wt ± >1000 157 ±9 317 ± 17 333 ± 55 567 ± 109 357 ± 102 313 ±61 187 ±9
Human Ovarian ADDP mt 12 ± 0.8 >1000 183 ±17 >1000 275 ± 56 260 ± 40 210 ±18 208 ± 19 233 ± 23
Human Osteosarcoma 143B mt 10.2 ±1.2 >1000 248 ± 29 665 ± 225 450 ± 50 >1000 327 ± 67 385 ± 43 223 ± 44
Human Colon HCT116 wt 12 ± >1000 160 ±10 >1000 78 +7 143 ± 23 180 ±20 173 ±22 107 ± 12
Human Colon HT29 mt 6.4 ± 0.7 >1000 183 ±20 530 ± 112 14 ± 0.3 28 ± 1 297 ± 58 373 ± 54 205 ± 13
Human Colon WiDr mt 6.1 ± 0.5 >1000 198 ± 53 620 ± 31 15±3 31 ± 10 320 ± 20 367 ± 44 190 ± 35
Human Colon SW480 mt 17.5 ± >1000 155 ±9 444 ± 27 88 ±5 247 ± 14 333 ± 22 353 ± 20 147 ± 14
Figure imgf000027_0001
|-
CΛ wt = wildtype, mt = mutant.
180-367μM) and MK-8 (IC50 173-385μM). Greater cytotoxicity was observed with MK-
2 (IC50 157-248μM) and MK-9 (IC50 107-233μM) which was also consistent across the
nine cell lines. The greatest cytotoxicity was observed with the MK-4 and MK-5
analogues, however, the magnitude of this response was cell line dependent. In this
context, MK-4 and MK-5 were selectively more cytotoxic in the human colon cancer
cell lines (IC50 14-88μM; 28-247μM) compared with leukaemia (IC50 393-680μM; 323-
>1000μM) ovarian (IC50 275-333μM; 260-567μM), and osteosarcoma (IC50 450μM;
>1000μM) cells respectively.
(b) Abrogation of cell cycle checkpoints:
The ability of the protein phosphatase inhibitors to abrogate the G, or G2
checkpoint of the cell cycle may be determined by cell cycle analysis using flow
cytometry. Briefly, asynchronous cell cultures are harvested 18h after 6Gy irradiation
and/or 12h incubation with the protein phosphatase inhibitor. Depending upon the p53
status of the cell line, radiation treatment alone will induce arrest in either G, and/or G2
phase of the cell cycle.
Data shown in Table 2 and Figure 4 show the cell cycle response of LI 210,
HL60, HT29 and HCTl 16 cells to cantharidin and the new cantharidin analogues MK-2
and MK-4 after 12h exposure. In summary, cantharidin and MK-2 produced a similar
response and induced G2 arrest in all four cell lines tested. MK-4 also induced G2 arrest
but only in L1210, HL60 and HCTl 16 cells. In HT29 cells, MK-2 induced G, cell cycle
arrest. The magnitude of the cell cycle arrest induced by these drugs directly correlated
with their cytotoxicity in the respective cell lines. The ability of the parent compound
cantharidin to inhibit cell growth is also shown (IC50 6.1-18μM). The cytotoxicity of the
SUBSTITUTE SHEET (RULE 26) Cell Cycle Analysis Table 2
Cell Cycle Distribution (percentage of total) of tumour cell lines 12h after cantharidin or cantharidin analogue treatment.
Method : Flow Cytometry of Propidium Iodide stained cells.
Agent L1210 cells HL60 cells HCTΪ 16 cells HT29 cells μM sub G, G, S G2+M sub G, G, S sub G, G, S G2+M sub G, G, S G2+M
Cantharidin 0 0.5 47.4 34.3 19.4 1.9 45.5 25.8 28.2 6.5 43.3 14.6 36.4 11.1 45.3 8.0 36.0
1 0.5 45.8 33.7 21.6 1.5 44.0 26.1 29.7 2.2 39.9 17.2 41.9 9.0 46.2 7.8 37.4
5 0.6 46.5 32.6 21.9 1.7 41.4 27.7 30.6 2.9 39.9 16.8 41.8 4.0 47.4 9.3 39.8
10 0.5 49.1 33.0 18.9 1.7 41.8 27.5 30.4 G2 arrest 6.2 38.0 14.9 42.0 2.8 42.7 14.6 40.3 G2 arrest
50 1.9 22.0 27.8 50.6 G2 arrest 19.3 16.2 31.6 34.7 Cell Death 11.1 25.1 17.8 48.1 G2 arrest 15.1 46.0 14.7 26.0 Cell Death
Figure imgf000029_0001
MK-2 0 0.4 40.1 28.4 32.1 2.1 45.6 21.0 32.6 4.7 44.2 13.7 36.8 6.0 46.4 9.3 37.5
^ 50 0.3 42.7 26.2 31.7 1.8 44.1 23.8 31.4 1.3 47.2 13.8 37.4 9.4 45.3 7.6 37.1 NJ
100 0.6 45.2 22.4 32.4 1.8 43.3 23.6 32.4 1.7 47.2 16.0 34.5 3.6 49.8 8.2 37.6 tn 250 2.4 46.7 14.3 36.7 3.2 37.7 23.8 36.4 G2 arrest 1.4 52.8 11.1 34.3 4.2 41.4 11.2 42.5 G2 arrest
CΛ 500 3.9 26.3 10.1 60.0 G2 arrest 18.8 17.8 21.6 43.1 Cell death 2.5 39.4 11.3 46.5 G2 arrest 5.2 44.5 15.4 33.6 S-phase
MK-4 0 0.8 42.0 26.9 31.7 2.3 49.9 21.6 27.4 4.1 44.0 12.5 39.4 5.5 45.7 7.4 41.4
50 0.5 42.0 26.9 32.0 1.9 44.7 22.3 32.3 4.5 43.9 11.4 40.7 4.7 51.4 12.3 31.6
100 0.4 43.2 25.4 32.5 2.5 45.3 22.6 30.6 2.0 41.4 13.6 44.1 6.0 52.3 12.5 29.4
250 0.5 45.7 24.6 30.5 6.0 40.0 23.0 32.0 3.9 36.2 14.1 46.9 7.0 53.2 11.9 27.6
500 1.1 47.5 18.6 33.9 Slight Δ 6.1 27.8 22.8 44.4 G2 arrest 9.6 29.0 15.7 46.5 G2 arrest 3.4 53.7 14.1 29.1 G, arrest
Figure imgf000029_0002
cantharidin is greater than for its analogues. Interestingly, cantharidin also showed slight
selectivity towards the colon cancer cells.
If the protein phosphatase inhibitor abrogates the G2 checkpoint then the cells
will not arrest in the G2 phase of the cell cycle and the cells will continue through the
cell cycle and accumulate in the Gi phase of the cell cycle only. Similarily if the protein
phosphatase inhibitors abrogates the G, checkpoint then the cells will not arrest in the Gj
phase of the cell cycle and accumulate in the G2 phase of the cell cycle only. Cell cycle
analysis using propidium iodide labelling of DNA has been used extensively in our
laboratory to assess the effect of specific anticancer agents that induce S-phase cell cycle
arrest and apoptotic cell death (Sakoff, Ackland and Stewart, 1998). Experiments were
performed on a Becton Dickinson FACScan and using Cell Quest software.
Data shown in Table 3 and Figure 5 show the cell cycle response of LI 210,
HL60, HT29 and HCTl 16 cells. The cells were treated with 6Gy of radiation and then
treated with cantharidin 6h later. The ability to abrogate cell cycle arrest was assessed
12h after the addition of the drugs. Cantharidin and MK-2 both abrogated radiation
induced G, arrest in all cell lines. MK-4 also abrogated G, arrest in L1210, HL60 and
HCTl 16 cells. In HT29 cells, MK-4 induced abrogation of the G2 checkpoint. It is
important to note that the exposure of HT29 cells to MK-4 induced the greatest
cytotoxicity (IC50 14μM) as determined by the MTT assay. Not surprisingly, the ability
to abrogate the G2 checkpoint was more lethal than the ability to abrogate the G!
checkpoint.
SUBSTITUTE SHEET (RULE 26) Table
Checkpoint Abrogation g
Cell Cycle Distribution (percentage of total) of tumour cell lines 18h after 6Gy of radiation and 12h after cantharidin or cantharidin analogue treatment.
Method : Flow Cytometry of Propidium Iodide stained cells.
Agent L1210 cells HL60 cells HCT116 cells HT29 cells μM sub G, G, S G2+M sub G G, S G,+M sub G G, S G2+M sub G G, S G2+M
Cantharidin 0 1.6 25.3 35.8 38.8 6.6 5.3 3.2 85.3 4.9 26.8 8.7 60.1 5.9 40.7 9.2 44.7
1 1.6 27.2 25.6 37.5 6.2 5.2 3.0 85.8 4.2 26.1 13.8 58.2 16.6 35.2 10.6 38.2
5 2.4 25.8 31.9 41.5 4.4 5.7 3.5 86.8 4.0 23.6 10.4 63.3 5.3 38.6 10.6 46.3
10 3.4 24.9 29.4 43.7 5.3 5.6 4.3 85.1 4.2 25.7 9.4 62.2 6.4 21.1 12.3 60.8
50 4.9 4.1 15.6 77.4 4.3 10.2 11.3 64.9 Cell Death 12.0 12.2 15.6 63.3 G, abrogation 14.7 23.0 20.7 43.1
Figure imgf000031_0001
MK-2 0 1.6 16.4 31.1 52.1 7.0 5.9 2.2 85.1 3.7 30.8 7.9 57.2 17.3 31.8 8.7 41.3
?α 50 4.0 19.0 27.8 50.1 5.9 6.1 2.8 85.5 3.3 32.8 6.3 57.3 10.3 35.4 8.8 44.7 c 100 3.5 18.4 23.0 55.8 5.5 6.1 3.3 85.4 3.1 29.9 7.2 59.6 3.5 40.6 9.0 45.9 tn 250 6.9 11.2 10.0 71.9 8.1 5.4 2.8 83.9 6.2 23.9 4.3 65.4 2.7 24.9 12.3 59.4
CΛ 500 5.4 3.4 2.9 88.4 1.8 4.4 4.1 80.0 Cell Death 6.4 15.4 4.9 73.0 G, abrogation 8.8 24.1 20.4 45.2
MK-4 0 1.9 20.2 29.7 50.0 8.7 5.7 2.0 83.9 10.3 31.4 6.2 52.1 7.0 35.1 9.3 48.4
50 1.8 21.2 28.5 50.3 8.9 6.2 2.8 82.3 6.3 26.7 5.8 61.3 6.8 28.9 16.4 48.4
100 2.4 22.0 27.4 49.7 9.8 6.2 3.4 80.8 3.3 18.4 9.7 69.3 6.3 33.3 17.2 43.3
250 3.1 21.2 24.6 52.7 9.3 5.8 3.1 82.2 8.2 16.3 8.2 67.8 10.3 35.2 17.3 36.5
500 5.0 18.2 16.0 61.8 G, abrogation 11.6 5.2 5.4 78.2 Cell Death 14.9 13.1 10.6 61.9 G, abrogation 3.9 39.4 19.2 37.7 G2 abrogation
(c) Combination studies:
The cell lines listed above are exposed continuously to cisplatin and the
phosphatase inhibitor in various drug ratio combinations for 72h and then assayed for
cytotoxicity. Similarly, the cells are exposed to 8 Gy of radiation and incubated with the
phosphatase inhibitor and assessed for cytotoxicity at 72 h.
Data shown in Figures 6-9 shows the results of combination studies utilising the
Median Effect Method in HT29 and HCTl 16 human colon cells. This method tests the
cytotoxicity of various drug combinations from which a combination index can be
calculated. A value of greater than one indicated antagonism, a value equal to 1 indicates
additivity, while a value less than one indicates synergism. The HT29 and HCTl 16 cell
lines were chosen as they have differing p53 status and they represent the tumour types
that responded the greatest to cantharidin and its analogues.
The data show that the simultaneous combination of cisplatin and MK-4 in both
HCTl 16 and HT29 cells was additive and not synergistic using drug molar ratios of 1 : 1 ,
10:1 and 1 : 10. An additive response indicated that the drugs were mediating their effects
via two separate biochemical pathways. The simultaneous combination of taxotere and
MK-4 in HT29 cells was also additive using drug molar ratios of 1 : 10, 1 : 100, 1 :1000
(Taxotere: MK-4). However, this drug combination of taxotere and MK-4 induced a
synergistic response in HCTl 16 cells. A synergistic response indicates that the two
drugs were interacting in such a way as to enhance the overall cytotoxic response and to
induce "more than the additive1' response of each individual agent. Consequently, the
addition of subtoxic levels of MK-4 clearly enhanced the cytotoxicity of taxotere.
SUBSTITUTE SHEET (RULE 26) Example 8
Results and Discussion
Anhydrides and simple analogues were synthesised according to literature
procedures (Eggelte et. al; 1973), and then subjected to a PPl and PP2A bio-assay (see
biochemistry) to determine their ability to inhibit these enzymes. The results of initial
screening at 100 mMs are shown in Table 4, along with IC50 values in some instances.
Table 4 The inhibition of protein phosphatase 1 and 2A by anhydride modified
cantharidin analogues.
Figure imgf000033_0001
SUBSTITUTE SHEET (RULE 26)
Figure imgf000034_0001
Of the compounds listed in Table 4, only 1 and 2 show any significant inhibition
of PP2A. at 97% and 95% respectively (with little selectivity apparent for either
enzyme). Interestingly the bioisoseteric replacement of the anhydride oxygen atom of 1
results in a complete loss of inhibition. Indeed no modification of the cyclic anhydride,
is tolerated, and consequently results in no inhibition of PP2 A.
Previously we have shown that analog 2 undergoes a rapid conversion to the
dicarboxylic acid under assay conditions. We thus examined the stability of the non-
SUBSTITUTE SHEET (RULE 26) active analogues (in Table 4) and found that they were stable under assay conditions
showing no decomposition, in fact 5 can be synthesised via the Diels-Alder reaction in water (Eggelte et al; 1973).
In all instances, the corresponding dicarboxylic acid derivatives display lower
inhibitory values at PP2A (Tables 5 and 6). Even though the anhydrides undergo a facile
ring opening to the dicarboxylic acids, the original conformation presented at the active
site must also play a role in determining the overall level of inhibition. Consequently,
we believe that the conformation of anhydride carbonyl groups is more favourable for
inhibition (essentially only one conformation presented at the active site), than that of the
dicarboxylic acid (four possible minimum energy conformations, data not shown).
Table 5 Effects of anhydride to dicarboxylic acid on the inhibition of PP2A
Figure imgf000035_0001
SUBSTITUTE SHEET (RULE 26)
Figure imgf000036_0002
In an attempt to determine the feasibility of anhydride opening via nucleophilic
attack from Tyr272, we conducted a series of model experiments in which 2 was allowed
to stand in a chloroform solution of phenol. This mixture was examined periodically by
Η NMR spectroscopy and showed the growth of a new species over a period of time (ca
10 days). Further analysis indicated the presence of a phenolate ester of norcantharidin
(scheme 4). Consequently, a metal assisted or nucleophilic attack under physiological
conditions represents a possible mode of assisted ring opening with the anhydride held in
a favourable conformation within the active site. In turn the resultant diacid rapidly
binds in a more favourable manner.
Figure imgf000036_0001
Scheme 4
SUBSTITUTE SHEET (RULE 26) Table 6 Inhibition of PPl and PP2A by selected cantharidin analogues
Figure imgf000037_0001
The results presented herein indicate that cantharidin analogues, via anhydride
opening are more potent inhibitors of PP2 A. Analogues in which the anhydride moiety
has been modified preventing a facile ring opening (except where otherwise indicated)
are extremely poor inhibitors of PP2A (Tables 5 and 6).
However, the most interesting result reported herein (see table 4) is the selective
inhibition of PPl by the dimethyl ester (3). Simple diesterification of 2 has completely
reversed the previously reported PP2A selectivity (ca 10 fold) of norcantharidin for
PP2A to yield selective small synthetic molecule for the inhibition of either PPl or
SUBSTITUTE SHEET (RULE 26) PP2A. Again this suggests that presentation of a diacid moiety to the active site is
crucial for the inhibition of PP2 A. No such restrictions are apparent with the limited structure activity data for PPl.
A synthetic inhibitor such as 3 represents a significant advance on the currently
widespread inhibitors of PPl and PP2A.
In conclusion, the present inventors have demonstrated that a facile ring opening
of the anhydride moiety is relevant for inhibition at PP2A. Also, that modification of the
dicarboxylic acid moiety gives rise to a PPl selective compound.
The above describes some embodiments of the present invention. Modifications
obvious to those skilled in the art can be made without departing from the scope of this
invention.
Industrial Applicability
It should be clear that the present invention will find light applicability,
especially in the medical and veterinary fields.
SUBSTITUTE SHEET (RULE 26) REFERENCES
Walsh A H, Chen A T, Honaken R E, FEBS Lett., 1997, 416, 230
Roberge M. Tudan C, Hung SMF, Flarder KW. Jirik FR, and Anderson H Cancer
Research 1994, 54, 61 15-6121.
McCluskey, A; Taylor, C; Quinn, R J; Suganuma, M; Fujiki, H; Bioorg. Med. Chem.
Lett., 1996, 6, 1025.
McCluskey A, Keane M A, Sim A T R, J Med. Chem. 1998, submitted.
Enz, A; Zenke. G; Pombo-Villar; Bioorg. Med. Chem. Lett., 1997, 7, 2513; and
Sodeoka, M; Baba, Y; Kobayashi, S; Hirukawa, N; Bioorg. Med. Chem. Lett., 1997, 7,
1833.
Cohen, P; Cohen, PTW; J. Biol Chem., 1989, 264, 21435.
Cohen, P; "77ze Structure and Regulation of Protein Phosphatases " Raven Press: New
York, 1990, p230-235.
Shenolinkar, S; Mairn, A C; Adv. Second Messenger Phosphoprotein Res., 1991b, 23, 1.
Krebs, E G; Angew. Chem.. Int. Ed. Engl., 1993, 32, 1122.
Fischer, E; Angew. Chem.. Int. Ed. Engl, 1992, 32, 1130.
Tachibana, K; Scheuer, P J; Tsukitani, Y; Kikuchi, H; Van Engen, D; Clardy, J;
Gopichand, Y; Schmitz, F J; J Am. Chem. Soc, 1981, 103, 2469.
Cheng, X-C; Kihara, T; Kusakabe, H; Magae, J; Kobayashi, Y; Fang, R-P; Ni, Z-F;
Shen, Y-C; Ko, K; Yamaguchi, I; Isono, K; J. Antibiot., 1987, 40, 907.
Botes, D P; Tuinman, A A; Wessels, P L; Viljoen, C C; Kruger, H; Williams, D H;
Santikarn, S; Smith, K J; Hammond, S J; J. Chem. Soc. Perkin Trans I, 1984, 2311.
SUBSTITUTE SHEET (RULE 26) Herada, K-I; Matsuura, K; Suzuki, M; Oka, H; Watanabe, M F; Oishi, S; Dahlem, A M;
Beasley, V R; Carmichael, W W; J. Chromatog., 1998, 448, 275.
Honaken, R E; FEBS Lett, 1993, 330, 283.
Matszawa, S; Suzuki, T; Suzuki, M; Matsuda, A; FEBS Lett., 1994, 356, 272.
Wang, G-S; J. Ethnoparmacol, 1989, 26, 147.
Cavill, G W K; Clark, D V; Naturally Occur ing Insecticides (M. Jacobson and D G
Crosby, EDS), Marcel Dekker, New York, 1971, 271.
Oaks, W W; DiTunno, J F; Magnani, T; Levey, H A; Mills, L C; A.M.A. Arch. Int.
Med., 1960, 105, 106.
Goldfarb, M T; Gupta, A K; Sawchuk, W S ; Dermatologic Clinics, 1991, 9, 287.
A. Schmitz, D G; J. Vet, Intern. Med., 1989, 3, 208.
Polettini, A; Crippa, U; Ravagii, A; Saragoni, A.; Forensic. Sci. Int., 1992, 56(1), 37.
Li, Y-M; Casida, J E; Proc. Nat I. Acad. Sci. USA, 1992, 89, 1 1867.
McCluskey, A; Taylor, C; Quinn, R J; Suganuma, M; Fujiki, H; Bioorg. Med. Chem.
lett., 1996, 6, 1025.
Eggelte, T A; de Koning, H; Huisman, H O; Tetrahedron, 1973, 29, 2445; 3 see:
Tamura, Y; Imanishi, K; Miki, Y; Ikeda, M; Synthesis, 1997, 559; 5 & 9 see: Kwart, H;
Burchuk. I; J. Am. Chem. Soc, 1952, 74, 3094.
Goldberg, J; Huang, H-B;; Kwon, Y-G; Greengard, P; Nairn, A C; Kuriayan, J; Nature,
1995, 376, 745.
Quinn, R J; Volter, K; Embury, K J, unpublished results.
SUBSTITUTE SHEET (RULE 26) Cohen, P; Alemany, S. ;Hemmings, B A; Resink, T J ; Stralfors, P and Tung, H Y L in
Methods in Enzymology (Ed. J D Corbin and R A Johnson) 1988, 159, 390, Academic
Press (London).
Hardie, DG; Haystead TAJ; Sim ATR; Protein Phosphorylation, in methods in
enymology, in press. Enz, A; Zenke, G; Pombo-Villar; Bioorg. Med. Chem. Lett.,
1997, 7, 2513.
Sodeoka, M; Baba, Y; Kobayashi, S; Hirukawa, N; Bioorg. Med. Chem. Lett., 1997, 7,
1833.
Durfee T.. et al., (1993). The retinoblastoma protein associates with the protein
phosphatase type 1 catalytic subunit. Genes Dev. 7, 555-569.
Ghosh, S. et al. (1996). Okadaic acid overrides the S-phase checkpoint and accelerates
progression of G2-phase to induce premature mitosis in Hela cells. Exp. Cell Res. 227:165-
169.
Herwig, S., and Strauss, M. (1997). The retinoblastoma protein: a master regulator of
cell cycle, differentiation and apoptosis. Eur.J.Biochem. 246:581-601.
Jackman, AL and Calvert. AH (1995). Folate-based thymidylate synthase inhibitors as
anticancer drugs. Ann.Oncol. 6:871-881, 1995.
Kawamura K-I, Grabowski D, Weizer K, Bukowski R, and Ganapathi R (1996).
Modulation of vinblastine cytotoxicity by dilantin (phentoin) or the protein phosphatase
inhibitor okadaic acid involves the potentiation of anti-mitotic effects and induction of
apoptosis in human tumour cells. Br. J. Cancer 73: 183-188.
Lazzereschi, D. et al. (1997). The phosphatase inhibitor okadaic acid stimualtes the
TSH-induced Gl-S phase transition in thyroid cells. Exp. Cell Res. 234:425-433.
SUBSTITUTE SHEET (RULE 26) Nakamura K. and Antoku S. (1994). Enhancement of X-ray cell killing in cultured
mammalian cells by the protein phosphatase inhibitor calyculin A. Cancer Res 54:2088-
20990.
O'Connor PM (1996). Cell cycle checkpoints: Targets for anti-cancer therapy. Anti-Cancer
Drugs 7(sup 3): 135-41.
O'Connor PM (1997). Mammalian GI and G2 phase chekpoints. Cancer Surveys 29:151-
182.
Peters GJ and Ackland SP (1996). New antimetabolites in preclinical and clinical
development. Exp. Opin. Invest. Drugs 5, 637-679.
Schsnthal A (1992). Okadaic acid - a valuable new tool for the study of signal
transduction and cell cycle regulation? New Biol. 4, 16.
Stein. G.S. et al. (1998). The molecular basis of cell cycle and growth control. Wiley-Liss
Publishers. New York.
Yamashita K.. et al (1990). Okadaic acid, a potent inhbitor of type 1 and type 2A protein
phosphatases, activates cdc2/Hl kinase and transiently induces a premature mitosis-like
state in BHK21 cells. EMBO J. 9: 4331-4338.
SUBSTITUTE SHEET (RULE 26)

Claims

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS :-
1. A cell permeable inhibitor of protein phosphatase, said inhibitor being an anhydride modified cantharidin analogue, ϊ
2. An inhibitor according to claim 1, wherein the phosphatase is phosphatase 1 and/or phosphatase 2A.
3. An inhibitor according to claim 1 or 2 wherein the anhydride modified cantharidin analogue is oxidatively stable.
4. A compound of the formula:
Figure imgf000043_0001
wherein R, and R2 are H. aryl or alkyl; X is O, N or S; Y is O, S, SR, NH, NR, CH2OH,
CH2OR; R is alkyl or aryl; A and B are H or CH3; W and Z are CHOH or C=0 and R,
and R2 can cyclise to form a ring as follows:
Figure imgf000043_0002
wherein R3 and R are H, aryl or alkyl.
5. A compound according to claim 3, wherein the aryl group is phenyl or naphthyl
and wherein the aryl group is attached via a carbon spacer of between 6 and 10 carbon
atoms.
6. A compound according to claim 3 or claim 4, wherein the alkyl group is C,-C10.
SUBSTITUTE SHEET (RULE 26)
7. A process for producing anhydride modified cantharidin analogues for use in the
treatment of cancer or for the sensitising cancer cells to one or more cancer treatments
comprising the step of reacting a diene with an ene.
8. A process according to claim 7 further comprising hydrogenation of the adduct
5 of the diene and the ene.
9. A process according to claim 7 or 8 further comprising ring opening of the
adduct of the diene and the ene.
10. A process for producing anhydride modified cantharidin analogues, said process
including the steps of:
o dissolving a diene in a suitable solvent and adding to the resultant solution an
ene.
11. A process for producing anhydride modified cantharidin analogue, said process
including the steps of:
dissolving a furan in a suitable solvent and adding to the resultant solution an
5 ene;
incubating the solution at a temperature and for a time sufficient to form a
precipitate; and
collecting the precipitate and recrystalising the analogue.
12. A process for producing anhydride modified cantharidin analogue, said process
0 including the steps of:
mixing thiophene and maleic anhydride at room temperature in a suitable
solvent;
SUBSTITUTE SHEET (RULE 26) compressing the mixture at a temperature and pressure sufficient to facilitate a reaction to take place; and
purifying the analogue.
13. A method of treating cancer which method comprises administering to a patient
in need of such treatment, an effective amount of an inhibitor according to any one of
claims 1 to 3 or a compound according to any one of claims 4 to 6, together with a
pharmaceutically acceptable carrier, diluent and/or excipient.
14. A method according to claim 13, wherein the cancer is inherently resistant to
conventional chemotherapy.
15. A method according to claim 13 or claim 14, wherein the cancer is colon cancer
or non small-cell lung cancer.
16. A method according to any one of claims 13 to 15, wherein the inhibitor or the
compound is administered intravenously.
17. A method of sensitising cancer cells to at least one method of treating cancer,
which method of sensitising comprises administering to a patient in need of such
treatment, an effective amount of an inhibitor according to any one of claims 1 to 3 or a
compound according to any one of claims 4 to 6, together with a pharmaceutically
acceptable carrier, diluent and/or excipient.
18. A method according to claim 17, wherein the at least one cancer treatment is
selected from treatments involving irradiation and anti-cancer agents.
19. A method according to claim 17 or claim 18, wherein the cells have deficient
p53 activity.
SUBSTITUTE SHEET (RULE 26)
20. A method of treating cancer which method comprises:
administering to a patient in need of such treatment, an effective amount of an
anhydride modified cantharidin analogue of any one of claims 1 to 3 or a compound
according to any one of claims 4 to 6 to sensitise cells of the patient to one or more
cancer treatments; and
utilising the one or more cancer treatments.
21. A method of screening compounds for use in sensitising cancer cells to at least
one method of treating cancer, and comprising:
screening for anti-cancer activity; and
screening for ability to abrogate either the Gt or the G2 checkpoint of the cancer
cell cycle.
22. A method according to claim 21 further comprising the step of screening for the
ability of the compounds of sensitise cancer cells to one or more cancer treatments.
23. A method according to claims 21 or 22 wherein the one or more cancer
treatments are selected from treatments involving cisplatin, irradiation, taxanes and
antimetabolites.
24. A method according to any one or claims 21 to 23 wherein the screening is
conducted on haematopoietic cells or solid tumour cells, having varying p53 activity.
25. A method according to claim 24, wherein the cells are selected form the group
consisting of L1210 (murine leukaemia, p53 wildtype), HL60 (human leukaemia, p53
nul), A2780 (human ovarian carcinoma, p53 wildtype), ADDP (cisplatin resistant
A2780 cells, p53 mutant), SW480 (human colon carcinoma, p53 mutant), WiDr (human
SUBSTITUTE SHEET (RULE 26) colon carcinoma, p53 mutant), HT29 (human colon carcinoma, p53 mutant), HCTl 16
(human colon carcinoma. p53 wildtype) and 143B (human osteosarcoma, p53 mutant).
26. A compound selected from a group comprising compounds (a) to (k) below:
(a) (b)
Figure imgf000047_0001
(c) ( )
Figure imgf000047_0002
(e) (f)
Figure imgf000047_0003
(g) (h)
Figure imgf000047_0004
(i) 0)
Figure imgf000047_0005
SUBSTITUTE SHEET (RULE 26) (k) (1)
Figure imgf000048_0001
(m) (n)
Figure imgf000048_0002
27. Use of an inhibitor according to claim 1 or claim 2, or a compound according to
any one of claims 3 to 5 for the manufacture of a medicament for the treatment of
cancer.
28. Use according to claim 27, wherein the cancer is colon cancer or non small-cell
lung cancer.
29. Use according to claim 27 or claim 28, wherein the medicament is administered
intravenously.
SUBSTITUTE SHEET (RULE 26)
PCT/AU1999/000567 1998-07-14 1999-07-14 Anhydride modified cantharidin analogues useful in the treatment of cancer WO2000004023A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2000560129A JP2002520415A (en) 1998-07-14 1999-07-14 Anhydrous modified cansalidin analogues useful for the treatment of cancer
KR1020017000638A KR20010074721A (en) 1998-07-14 1999-07-14 Anhydride modified cantharidin analogues useful in the treatment of cancer
AU47634/99A AU772501B2 (en) 1998-07-14 1999-07-14 Anhydride modified cantharidin analogues useful in the treatment of cancer
EP99930941A EP1104423A4 (en) 1998-07-14 1999-07-14 Anhydride modified cantharidin analogues useful in the treatment of cancer
CA002337771A CA2337771A1 (en) 1998-07-14 1999-07-14 Anhydride modified cantharidin analogues useful in the treatment of cancer
AU2004203476A AU2004203476A1 (en) 1998-07-14 2004-07-29 Anhydride modified cantharidin analogues useful in the treatment of cancer
US11/240,971 US20060030616A1 (en) 1998-07-14 2005-09-30 Anhydride modified cantharidin analogues useful in the treatment of cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AUPP4665A AUPP466598A0 (en) 1998-07-14 1998-07-14 Product and process
AUPP4665 1998-07-14

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09743841 A-371-Of-International 2001-05-09
US10/703,336 Continuation US20040110822A1 (en) 1998-07-14 2003-11-07 Anhydride modified cantharidin analogues useful in the treatment of cancer

Publications (2)

Publication Number Publication Date
WO2000004023A1 WO2000004023A1 (en) 2000-01-27
WO2000004023A9 true WO2000004023A9 (en) 2000-04-20

Family

ID=3808903

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU1999/000567 WO2000004023A1 (en) 1998-07-14 1999-07-14 Anhydride modified cantharidin analogues useful in the treatment of cancer

Country Status (6)

Country Link
EP (1) EP1104423A4 (en)
JP (1) JP2002520415A (en)
KR (1) KR20010074721A (en)
AU (1) AUPP466598A0 (en)
CA (1) CA2337771A1 (en)
WO (1) WO2000004023A1 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10038043B4 (en) * 2000-08-02 2006-09-07 Walter, Michael, Dr. Phamacologically active substance for the treatment of cardiovascular diseases
AUPR392301A0 (en) * 2001-03-23 2001-04-26 University Of Newcastle Research Associates Limited, The Protein phosphatase inhibitors
JP2004532260A (en) * 2001-05-31 2004-10-21 ザ チャイニーズ ユニバーシティ オブ ホンコン Composition containing demethylcantharidin in combination with a platinum-containing anticancer agent and use thereof
AU2003270218B2 (en) 2002-10-02 2008-11-20 Sanofi-Aventis Deutschland Gmbh Hydroxyphenylundecane derivatives, a process for their production and their use
CN1623993A (en) * 2003-12-02 2005-06-08 北京键凯科技有限公司 Cantharides amine and demethyl cantharides amine derivative and application in medicine thereof
CN1304396C (en) * 2005-04-08 2007-03-14 中山大学 Derivative of canthardin and preparation method
US8299076B2 (en) 2005-05-18 2012-10-30 Array Biopharma Inc. Crystalline forms of 2-(2-flouro-4-iodophenylamino)-N-(2-hydroxyethoxy)-1,5-dimethyl-6-oxo-1,6-dihydropyridine-3-carboxamide
EP1736154A1 (en) * 2005-06-24 2006-12-27 Friedrich-Alexander-Universität Erlangen-Nürnberg Use of PP-1 inhibitors to prevent missplicing events
CN103788108B (en) * 2007-02-06 2017-04-12 利克斯特生物技术公司 Oxabicycloheptanes and oxabicycloheptenes, their preparation and use
US8227473B2 (en) 2008-08-01 2012-07-24 Lixte Biotechnology, Inc. Oxabicycloheptanes and oxabicycloheptenes, their preparation and use
AU2009277086B2 (en) 2008-08-01 2015-12-10 Lixte Biotechnology, Inc. Neuroprotective agents for the prevention and treatment of neurodegenerative diseases
WO2010147612A1 (en) 2009-06-18 2010-12-23 Lixte Biotechnology, Inc. Methods of modulating cell regulation by inhibiting p53
AU2014251087B2 (en) 2013-04-09 2019-05-02 Lixte Biotechnology, Inc. Formulations of oxabicycloheptanes and oxabicycloheptenes
KR101511857B1 (en) * 2013-11-01 2015-04-13 주식회사 엘지생활건강 A body hair growth inhibition composition comprising dimethyl cantharidin as an effective ingredient
CN104447782B (en) * 2014-12-30 2016-09-14 贵州柏强制药有限公司 Bromo norcantharidin mono-acid benzyl ester and synthetic method thereof and application
KR102296703B1 (en) 2017-03-20 2021-09-01 포르마 세라퓨틱스 인크. Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators
BR112021005188A2 (en) 2018-09-19 2021-06-08 Forma Therapeutics, Inc. treating sickle cell anemia with a pyruvate kinase r activating compound
US20230055923A1 (en) 2018-09-19 2023-02-23 Forma Therapeutics, Inc. Activating pyruvate kinase r
KR20240012182A (en) 2022-07-20 2024-01-29 고려대학교 산학협력단 Composition comprising the cantharidine compound for prevention or treatment of glioblastoma

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3954913A (en) * 1975-01-06 1976-05-04 The Standard Oil Company Stabilized nitrile polymers
US4220594A (en) * 1977-11-04 1980-09-02 E. R. Squibb & Sons, Inc. Hexa- and octahydro-4,7-epoxyisobenzofuran-1-ol and hexa- and octahydro-5,8-epoxy-1H-2-benzopyran-3-ol
US4228180A (en) * 1979-11-01 1980-10-14 E. R. Squibb & Sons, Inc. 7-Oxabicycloheptane and 7-oxabicycloheptene prostaglandin analogs
JPS63170383A (en) * 1987-02-26 1988-07-14 Nippon Zeon Co Ltd Alicyclic dicarboximide compound
DE4134755A1 (en) * 1991-10-22 1993-04-29 Bayer Ag USE OF PARTLY KNOWN SUBSTITUTED TETRAHYDROTHIOPHENES AS A MEDICINAL PRODUCT, NEW ACTIVE SUBSTANCES AND METHOD FOR THE PRODUCTION THEREOF
DE4134758A1 (en) * 1991-10-22 1993-04-29 Bayer Ag NEW SUBSTITUTED TETRAHYDROFURANE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A MEDICINAL PRODUCT
US5399725A (en) * 1993-05-27 1995-03-21 Bristol-Myers Squibb Co. 7-oxabicycloheptane carboxylic acid prostaglandin analog intermediates useful in the preparation of anti-thrombotic and anti-vasospastic compounds and method for preparing same
WO1995007022A1 (en) * 1993-09-07 1995-03-16 Zeneca Limited Fungicides
CA2180260A1 (en) * 1993-12-29 1995-07-06 Dennis M. Brown Methods and compositions for the treatment of a host with a cellular proliferative disease

Also Published As

Publication number Publication date
WO2000004023A1 (en) 2000-01-27
CA2337771A1 (en) 2000-01-27
KR20010074721A (en) 2001-08-09
AUPP466598A0 (en) 1998-08-06
JP2002520415A (en) 2002-07-09
EP1104423A4 (en) 2001-10-24
EP1104423A1 (en) 2001-06-06

Similar Documents

Publication Publication Date Title
US20060030616A1 (en) Anhydride modified cantharidin analogues useful in the treatment of cancer
WO2000004023A9 (en) Anhydride modified cantharidin analogues useful in the treatment of cancer
Li et al. Asperversiamides, linearly fused prenylated indole alkaloids from the marine-derived fungus Aspergillus versicolor
Song et al. β-Carbolines as specific inhibitors of cyclin-Dependent kinases
JP5926727B2 (en) Substituted imidazo [1,2-b] pyridazine
Shi et al. Design, synthesis and in vitro and in vivo antitumor activities of novel bivalent β-carbolines
Zhang et al. Synthesis and structure–activity relationships of N2-alkylated quaternary β-carbolines as novel antitumor agents
HUE034591T2 (en) Selective pi3k delta inhibitors
Shi et al. Antitumor agents. 172. Synthesis and biological evaluation of novel deacetamidothiocolchicin-7-ols and ester analogs as antitubulin agents
Xu et al. 17β-Hydroxywithanolides as sensitizers of renal carcinoma cells to tumor necrosis factor-α related apoptosis inducing ligand (TRAIL) mediated apoptosis: Structure–activity relationships
JPH08176070A (en) Didepside derivative and pi3 kinase inhibitor
Bihud et al. Goniolanceolatins A–H, cytotoxic Bis-styryllactones from Goniothalamus lanceolatus
CN110156822B (en) Naphthol-phenylboronic acid compound and preparation method and application thereof
US9301956B2 (en) Application of cinchona alkaloid derivatives as cytotoxic compounds
Wang et al. Meroterpenoids with a large conjugated system from Ganoderma lucidum and their inhibitory activities against renal fibrosis
US9399644B2 (en) [1,3] dioxolo [4,5-G] quinoline-6(5H)thione derivatives as inhibitors of the late SV40 factor (LSF) for use in treating cancer
Liu et al. Retracted Article: Synthesis of xanthone derivatives and anti-hepatocellular carcinoma potency evaluation: induced apoptosis
AU772501B2 (en) Anhydride modified cantharidin analogues useful in the treatment of cancer
EP1200444A1 (en) Novel xanthone compounds, their preparation and use as medicament
Ayogu et al. Phenothiazine derivatives as potential antiproliferative agents: A mini-review
AU2004203476A1 (en) Anhydride modified cantharidin analogues useful in the treatment of cancer
Dai et al. Synthesis and antitumor effect of sophoridine derivatives bearing an acyclic aryloxy phosphoramidate mustard functionality
JPH10203977A (en) Antitumor agent
US9682947B2 (en) Substituted lactones for treating cancer
US4874767A (en) Discorhabdin D, compositions containing same and methods of preparatio and use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: C2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C2

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

COP Corrected version of pamphlet

Free format text: PAGES 1-39, DESCRIPTION, REPLACED BY NEW PAGES 1-40; PAGES 40-45, CLAIMS, REPLACED BY NEW PAGES 41-46; PAGES 1/27-27/27, DRAWINGS, REPLACED BY NEW PAGES 1/27-27/27; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

ENP Entry into the national phase

Ref document number: 2337771

Country of ref document: CA

Kind code of ref document: A

Ref document number: 2337771

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1020017000638

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1999930941

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 47634/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 09743841

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1999930941

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020017000638

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: CA

WWW Wipo information: withdrawn in national office

Ref document number: 1020017000638

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 47634/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1999930941

Country of ref document: EP