AU721950B2 - Potentiation of temozolomide in human tumour cells - Google Patents

Potentiation of temozolomide in human tumour cells Download PDF

Info

Publication number
AU721950B2
AU721950B2 AU63711/98A AU6371198A AU721950B2 AU 721950 B2 AU721950 B2 AU 721950B2 AU 63711/98 A AU63711/98 A AU 63711/98A AU 6371198 A AU6371198 A AU 6371198A AU 721950 B2 AU721950 B2 AU 721950B2
Authority
AU
Australia
Prior art keywords
temozolomide
atase
inhibitor
administered
cells
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.)
Ceased
Application number
AU63711/98A
Other versions
AU6371198A (en
Inventor
John Colin Baer
Azadeh Alison Freeman
Geoffrey Paul Margison
Edward Stuart Newlands
Joseph Anthony Rafferty
Amanda Jean Watson
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.)
Cancer Research Campaign Technology Ltd
Original Assignee
Cancer Research Campaign Technology Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU58384/94A external-priority patent/AU5838494A/en
Application filed by Cancer Research Campaign Technology Ltd filed Critical Cancer Research Campaign Technology Ltd
Priority to AU63711/98A priority Critical patent/AU721950B2/en
Publication of AU6371198A publication Critical patent/AU6371198A/en
Application granted granted Critical
Publication of AU721950B2 publication Critical patent/AU721950B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

A
P/00/011 Regulation 3.2
AUSTRALIA
Patents Act 1990
ORIGINAL
COMPLETE SPECIFICATION STANDARD PATENT Invention Title: "Potentiation of Temozolomide in Human Tumour Cells" The following statement is a full description of this invention, including the best method of performing it known to us: GH REF: P23908C/TPG:EM I I WO 94/15615 PCT/GB94/00065
IA
POTENTIATION OF TEMOZOLOMIDE IN HUMAN TUMOUR CELLS BACKGROUND OF THE INVENTION Temozolomide, or 8-carbamoyl-3-methylimidazo[5,1-d]- 1,2,3,5,-tetrazin-4-(3H)-one, (CCRG 81045, NSC 362856) has been found to possess valuable antitumour properties, see Newlands et al., Br. J. Cancer, 65:287 (1992). In the clinic, temozolomide has shown activity against astrocytoma, gliomas, malignant melanoma and mycosis fungoides. The drug is most useful when administered according to a repeat dose schedule.
Methylated O'-alkylguanine, from reaction with MTIC (the active methylating species of temozolomide), is repaired by the protein O'-Alkylguanine
DNA
alkyltransferase (ATase). Pretreatment of ATase- 15 expressing cells with methylating agents SZlotogoski et al., Carcinogenesis, 5:83, 1984; Gibson et el., Cancer Res., 46:4995, 1986), O'-methylguanine Dolan et al., Biophys. Res. Commun., 132:178 1985) or 0'benzylguanine Dolan et al., Proc. Natl. Acad. Sci.
87:5368, 1990) has thus been shown to increase the cytotoxic effects of chloroethylating agents whilst little or no sensitization was observed in cells that do not express ATase.
Moschel, Dolan and Pegg, in U.S. Patent 5,091,430, note 25 that a transient decrease in ATase activity is all that is needed to enhance the effectiveness of chloroethylating agents. PCT published Application WO 91/13898 notes, for instance, a 3.8 fold decrease in the EDo for Me CCNU when combined with o'-benzylguanine in SF767 cells. Thus, Moschel et al. show a general enhancement of the anti-neoplastic activity .of an alkylating agent when used with a depletor of alkyltransferase.
Applicants' invention, which is surprising and unobvious in view of the earlier work, is that the chemotherapeutic effects of temozolomide can be dramatically potentiated (up to 300-fold for the MAWI cell line) by utilizing a particular dosing regimen which incorporates the administration of an ATase inhibitor. Thus, human cell cancers which were heretofore insusceptible or only mildly susceptible to temozolomide therapy can be treated by the combination of temozolomide with an ATase inhibitor.
The present invention relates to compositions and methods for improving and extending the therapeutic usefulness of temozolomide as an antineoplastic agent by a combination 15 therapy with a potentiator which is an inhibitor of the enzyme 0 6 -alkylguanine DNA alkyltransferase (ATase).
The present invention also relates to therapeutic regimens using these compositions and methods for the optimal potentiation of toxicity of 20 temozolomide to human cancer cells.
The present invention also relates to a repeat dosing regimen of temozolomide which is potentiated by prior or concomitant administration of an ATase inhibitor.
The present invention also relates to a method of determining the relative potentiation of temozolomide toxicity by an ATase inhibitor of a particular human cancer cell by ascertaining the amount of ATase produced by said cancer cell.
2A SUMMARY OF THE INVENTION In a first aspect the invention provides a method of potentiating the toxicity of temozolomide in human cancer cells by administering to a patient in need of such therapy an ATase inhibiting amount of an ATase inhibitor and an effective amount of temozolomide.
In a second aspect the invention provides a pharmaceutical composition, comprising an effective amount of an ATase inhibitor and an effective amount of temozolomide, when used for treating human cancer cells in a patient in need of such treatment.
In a third aspect the invention provides use of temozolomide and an ATase inhibitor for the manufacture of a pharmaceutical composition for the treatment of human cancer cells in a patient.
20 In a fourth aspect, the invention provides a kit when used in the method of the first aspect of the invention, the kit comprising a dosage form of temozolomide and a separate dosage form of an ATase inhibitor.
25 In a fifth aspect, the invention provides a product comprising temozolomide and an ATase inhibitor as a combined preparation when used for separate or sequential .administration to a patient in need of such treatment in repeat dosages on consecutive days in the treatment of human cancer cells.
DOC
3 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a graph of the cytotoxicity (ICs) of temozolomide and CCNU versus cellular Atase levels in the human tumour cell lines (in order of increasing ATase levels): ZR-75-1, U87MG, U373, LS174T, LOVO, MCF-7 and MAWI.
Figure 2 is a graph of the cytotoxicity of temozolomide in pZipneoSV(X)l-transfected (o,m or phAT-transfected (Y,A)XP-derived cell lines in the presence or absence of 10 pM BG. Error bars indicate 1 s.d.
Figure 3 is a graph of the cytoxicity ratio of repeated 20 daily doses of temozolomide in MAWI MCF-7 or U373 human tumour cell lines of drug only, IC, 0 compared to preincubation with BG, IC, Figure 4 is a graph of the effect of increasing concentrations of temozolomide on ATase levels in the human tumour cell lines: LOVO MAWI MCF-7 U373 Figure 5 is a graph of uptake of radiolabel at 4 0 C by cells treated with 1"C-temozolomide. MAWI ZR-75-1 In the claims which follow and in the preceding summary of the invention, except where the context requires otherwise due to express language or necessary implication, the word "comprising" is used in the sense of "including", i.e. the features specified may be associated with further features in various embodiments RT of the invention.
WO 94/15615 PCT/GB94/0UU06 4 DETAILED DESCRIPTION OF THE INVENTION The toxicity of temozolomide, an antitumour agent useful in the treatment of human cancers, can be greatly enhanced by its use in conjunction with a potentiator which is an inhibitor of the enzyme O'-alkylguanine
DNA
alkyltransferase (ATase). More particularly, the use of ATase inhibitors, such as O'-benzylguanine can enhance the toxicity of temozolomide, in, for example, the MAWI cell line, by up to about 300 fold when utilized in the scheduled dosage regimen of the present invention and enable the use of temozolomide in the chemotherapy of human cancers heretofore insusceptible or only mildly susceptible to such therapy.
A parallel toxicity for temozolomide and lomustine
(CCNU)
(after 1 hour drug exposure), can be shown with a number of human tumour cell lines, correlating with -their ATase content. Methylation of the O'-position of guanine in DNA by temozolomide thus results in a cytotoxic lesion. By screening human cancer cells for their relative ATase 20 production, sensitivity to temozolomide can be determined. A cell producing high amounts of ATase will thus be less sensitive to temozolomide alone than one producing minimal ATase levels.
Pretreating cells with a single dose of BG causes a modest fold) increase in temozolomide toxicity. The degree of enhancement for temozolomide and lomustine (CCNU) are a similar order of magnitude. In a colony assay, human ATase cDNA-transfected fibroblasts pretreated with BG remained more resistant to temozolomide than control transfected fibroblasts, although the ATase protein was eliminated. This is unlikely to be due to differences in temozolomide transport and may simply reflect resynthesis of ATase by WO 94/15615 PCTIGB94/00065 the phAT fibroblasts to diminish the effect of pretreatment with the inhibitor.
However, most surprisingly, a major potentiation of temozolomide toxicity (up to about 300 fold) by BG has been found in the MAWI cell line after five days treatment. A similar degree of enhancement was seen in MCF-7 cells which also contain high levels of ATase, but only a small effect in U373 cells which have low levels.
The results in the testing of the MAWI and MCF-7 cell lines imply that the continued presence of the ATase inhibitor permits a build up of DNA damage.
The present invention thus provides a method of potentiating the toxicity pf temozolomide in human cancer cells by administering an ATase inhibiting amount of an ATase inhibitor, and a product comprising temozolomide and an ATase inhibitor as a combined preparation for simultaneous, separate or sequential administration in said treatment of human cancer cells.
Preferably, this administration of an ATase inhibitor is 20 repeated over a period of several or multiple days, and is prior to the administration of the doses of temozolomide. Repeat doses can be administered at 1, 2, 3, 4 or 5 days, with 4 or 5 days being the preferred period of therapy.
25 Further, and most preferably, the temozolomide is administered to a patient in repeat doses over a period of days, and an ATase inhibiting amount of an ATase inhibitor is administered prior to each dose of temozolomide, resulting in a markedly increased toxicity of the temozolomide to human cancer cells, about 300 fold for the MAWI cell line.
The ATase inhibitor is administered in an ATase inhibiting amount, an amount sufficient to sensitize the tumour in vivo without causing undue sensitization of normal tissue, when the ATase inhibitor is used concurrently with temozolomide.
The amount of ATase inhibitor employed in the present invention to be used varies according to the degree of the effective amount required for treating tumour cells.
A suitable dosage is that which will result in a concentration of the ATase inhibitor in the tumour cells to be treated which results in the depletion of the ATase activity, about 1-2000 mg/kg, and preferably about 10-800 mg/kg, prior to chemotherapy.
The neoplasms for which temozolomide is a particularly suitable treatment include carcinomas, melanomas, sarcomas, lymphomas and leukaemias, with specific utility for astrocytoma, gliomas, malignant melanoma, and mycosis fungoides, Ewings sarcoma, chronic lymphocytic leukaemia, 20 and lung and breast tumours. Particularly dramatic enhancement of the temozolomide activity with an ATase Se inhibitor is found in breast, astrocytoma and colorectal tumour cells.
Typical dosage ranges of temozolomide are generally 25 between 0.1 and 200, preferably between 1 and 20, mg/kg body weight per day, or expressed in terms of body surface area, about 40-400, and preferably about 150-300 mg/m 2 per day.
The amount of potentiation by the ATase inhibitor is dependent upon the amount of ATase normally present in the particular cancer cell type. A cancer cell having higher levels of ATase will be potentiated more dramatically by the preadministration of the ATase R inhibitor.
WO 94/15615 PCTIGB94/00065 7 The ATase inhibitor utilizable in the present invention are those known to possess such activity, for instance, the o'-alkylguanines such as O'-methylguanine, the alkenylguanines such as O'-allylguanine and the 06arylguanines, such as O'-benzylguanine and the 06benzylated guanine, guanosine and 2'-deoxyguanosine compounds described in PCT International Application
WO
91/13898 (published September 19, 1991). Particularly suitable for use in the present invention is O6benzylguanine.
The particular dosage of ATase inhibitor depends upon the' amount of ATase normally found in the cancer cell being treated, the age and condition of the patient, and the particular ATase inhibitor being utilized.
Temozolomide has been found to be most preferably administered in repeat dosages on consecutive days, and the dramatic potentiation effects of the present invention are realized in the. highly preferred regimen involving the administration of an ATase inhibiting dose of the ATase inhibitor prior to, or concurrent with, the administration of each dose of temozolomide administered.
Preferably, 0'-benzylguanine is utilized as the ATase inhibitor, along with temozolomide administered at a daily rate of 150-300 mgm 2 in four or five divided doses over four or five consecutive days (total dose 750-1500 mg/m 2 Preferably, each dose of the ATase inhibitor is administered 2-8 hours prior to each dose of the o *temozolomide. This affords the greatest potentiation of temozolomide toxicity, and results in the most effective treatment of the patient's particular neoplasm. Most preferably, this regime is repeated after an interval of about four weeks.
An alternate dosage schedule for the administration of temozolomide with O'-benzylguanine is a continuous- VWO 94/15615 PCTIGB94/00065 8 schedule wherein the two drugs are administered on a daily basis for a period of four or more days. This combination therapy can be extended as needed on a continual basis until remission is attained.
Additionally, the ATase production of a particular human cancer cell can be utilized as a screening method to determine potentiation of temozolomide toxicity to said cell. In a preferred method, the ATase content of the particular cell line is assayed, by the method described by Lee et al., Cancer Res., 51:619 (1991), and the potential sensitivity to temozolomide determined.
Such a determination then enables the appropriate combination of temozolomide and ATase inhibition to be administered in a therapeutic regimen.
For preparing pharmaceutical compositions from the compounds described by this invention, inert, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets and 20 suppositories. The powders and tablets may be comprised of from about 5 to about 70 percent active ingredient.
Suitable solid carriers are known in the art, e.g., magnesium carbonate, magnesium stearate, talc, sugar, lactose. Tablets, powders, cachets and capsules can be used as solid dosage forms suitable for oral administration.
For preparing suppositories, a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted, and the active ingredient is dispersed homogeneously therein as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool and thereby solidify.
WO,94/15615 PCTIGB94/00065 9 Liquid form preparations include solutions, suspensions and emulsions. As an example may be mentioned water or water-propylene glycol solutions for parenteral injection.
Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.
The compounds of the invention may also be deliverable transdermally. The transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
Preferably, the pharmaceutical preparation is in unit dosage form. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component, an effective amount to achieve 20 the desired purpose..
The quantity of active compound in a unit dose of preparation may be varied or adjusted from about 0.1 mg to 1000 mg, more preferably from about 1 mg to 500 mg, according to the particular application.
25 The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired.
WO 94/15615 PCTIGB94/00065 Temozolomide may be administered using conventional techniques such as those described in Wasserman et al., Cancer, 36:1258-1268 (1975). Where appropriate, oral administration at a rate of 40-400 mgm- per day, and preferably 150-300 mgm4 2 per day, in 1-5, and preferably doses, over 1-5, and preferably 4-5, consecutive days is highly preferred. Intravenous administration at a daily dose of 25-250 mgm 2 is preferable for a continuous dosing therapy regimen. Oral administration can be utilized for a repeat dosing regimen.
The ATase inhibitor can be administered separately prior to, or concurrent with, the temozolomide. Where it is desirable to do so, both the ATase inhibitor and temozolomide can be combined into a unit dosage form to facilitate patient dosing. Such combination dosage forms may be in any of the above-described dosage forms, but, as noted above, are preferably in oral or intravenous forms.
o The temozolomide and the ATase inhibitor can be packaged 20 in a kit form. In such a kit, the temozolomide and the ATase inhibitor would be individually formulated into particular dosage forms for the particular route of administration, and contain instructions for the administration of the contents. In a typical embodiment for oral formulation, such a kit may be in the form of a blister package with separately formulated oral dosage forms of the temozolomide and the ATase inhibitor.
Any necessary adjustments in dose can be readily made to meet the chemotherapeutic treatment requirements of the individual patient and adjusted accordingly by the skilled practitioner.
WO'94/15615 PCT/GB94100065 11 The invention disclosed herein is exemplified by the following preparative examples, which should not be construed to limit the scope of the disclosure.
EXAMPLE 1 Materials and Methods Materials Tissue culture medium was purchased from ICN Biomedicals Ltd. (High Wycombe, UK) and fetal calf serum from Gibco Ltd (Paisley, UK). O'-benzylguanine (BG) was kindly supplied by Dr. R.C. Moschel (NCI-Frederick Cancer Research Development Center, Frederick, Maryland, USA).
Temozolomide and its chloroethyl analogue,-mitozolomide (8-carbamoyl-3-(2-chloroethyl)imidazo[5,1-d]-1, 2 tetrazin-4-(3H)-one), were synthesized by May and Baker Ltd (Dagenham, UK) and stored as solutions in DMSO at -700C. All other chemicals were purchased from Sigma Chemical Co. Ltd. (Poole, UK).
Ctotoxicity studies Cell lines were routinely grown as monolayers in DMEM 20 supplemented with 10% foetal calf serum, 25mM HEPES, glutamine and penicillin/streptomycin. Cytotoxicity studies were carried out in HEPES-free medium in a 5% CO, 2 atmosphere. 750-1000 cells/well were plated in 96 well plates and, after overnight incubation, were treated for 2 hours with or without 33 pM BG. Temozolomide or CCNU was then added for 1 hour in the same medium, the final DMSO concentration not exceeding The cells were grown for a further 7 days in fresh medium and assayed for protein content by NCI sulphorhodamine assay described by Skehan et al., J. Natl. Cancer Inst., _2:1107 (1990); growth studies showed that cells were in log phase growth during the assay period. For the repeat temozolomide dosing schedule, cells were given *WO 94/15615 PCT/GB94/00065 12 consecutive 24 hour treatments, with fresh medium each day. Assays were carried out at least in duplicate.
Human ATase cDNA-transfected or control XP cells [Fan et al., Nucleic Acid. Res., 1:5723 (1990)] were grown in MEM and 1000 cells/well were plated. After a 3 hour incubation, temozolomide, freshly diluted into MEM, was added and the plates incubated for 5 days. Survivals were assayed as described by Morten et al., Carcinogenesis, 13:483 (1992). In the BG experiments, 300 cells were plated in triplicate onto 9cm plates and allowed to attach for 5 hours. BG was added (10 pM in MEM) 3 hours prior to the treatment with temozolomide which was freshly diluted into MEM containing 10 pM BG.
After 7 days, colonies were strained with Giemsa and counted.
O'-alkvlcuanine DNA alkyltransferase assay This assay was carried out as described by Lee et al., Cancer Res., 51:619 (1991). Thus, varying amounts of cell extracts are incubated with DNA which contains 06- 20 methylguanine labelled with 3 H] in the methyl group, at preferably 37"C for 2 hours in a total volume of 300 pl of 1 mg/ml of bovine serum albumin in buffer 1. After incubation, bovine serum albumin (100pl of a 10 mg/ml solution in buffer 1) and perchloric acid (100pl of a 4 M 25 solution) are added in rapid succession. A further 2 ml of 1M perchloric is added and the mixture heated at for 40 minutes to degrade' the DNA to acid soluble material. The protein, which contains the methylated ATase, is then collected by centrifugation, washed with 4ml of 1M perchloric acid before being resuspended in 300 1l of 0.01M sodium hydroxide and dissolved in 3ml of aqueous scintillation fluid (Ecoscint A: National Diagnostics), and counted. The protein content of the cells was determined with a BioRad protein assay kit using bovine serum albumin as a standard. ATase activity WO 94/15615 PCTIGB94/00065 13 is expressed as fmol methyl transferred to protein per mg of total protein in the extract.
Cellular uptake of [4C] labelled temozolomide 8-carbamoyl-3- methylimidazo l-d -1,2,3,5-tetrazin- 4-(3H)-one (specific activity 26.3mCi/mmole) was kindly supplied by Dr. John Slack (Aston Molecules Ltd, Birmingham, UK). Cell suspensions (5 x 106/ml) were equilibrated at 40C and treated with 200 AM of the labelled drug. 10' cells were pipetted into eppendorf tubes and centrifuged through 250ul of an oil mixture (4:1 "Three-in-One"/Dow Corning silicone oil). The aqueous layer was aspirated and the oil layer gently washed with a further 300 1l of saline. After centrifugation both layers were aspirated, the cell pellet dissolved in a tissue solubilizer such as Protosol® (quaternary ammonium hydroxide in toluene) and added to scintillation vials containing Optiphase® •99% diisopropylnaphthalene).
000S :*0 Results of CvtotoxicitY studies The data are given below in Table I.
S
WO 94/15615 W094/5615PCTGB94OOO65 Single Dose Cvtotoxicitv CELL TEMOZOLOMIDE CCNUJ ATase.
LINE (fmollmg protein) 0
IC,
0 Ratio' IC50 IC50 Ratio'
[+BG]
(pM)M) (jd)
BREAST
ZR-75-1 32 23 1.4 12 25 0.5 MCF-7 325 171 1.9 70 31 2.2 1581.3
ASTRO-
CYTOMA
U87MG 172 131 1.3 28 8.8 3.2 21.9 U373 131 78 1.7 15 12 1.2 53.2
REML
LS 174T 873 632 1.4 73 13 5.7 199.6 LOVO 848 323 2.6 92 32 2.9 529.0 MAWI 1173 335 3.5 133 30 4.4 992.3 XP JNME pZip phAT 232 1 (0.8)2,3 <2 1002 1240L- Cells were exposed in tissue culture to /-06_ benzylguanine (BG) prior to a single dose of Temozolomide or CCNU.
1 IC 30 (-BGJ/1C 30
(+BGJ.
2. Results obtained by MTT assay of Wasserman et al., Int. J. Radiat. Oncol. Biol. Phvs.- 1,:699 (1988).
3. Figures in parentheses refer to mitozolomide.
The data of Table I which are graphically displayed in Figure 1 show a reasonable correlation between the sensitivity (as measured by the concentration which gives 50% of growth or.IC 0 of tumour cell lines to temozolomide (r correlation coefficient 0.87) or -CCNU (r correlation .coefficient 0.92) and their ATase .9 0* WO 94/15615 PCT/GB94100065 content. The slopes are nearly parallel except that CCNU is approximately five times more toxic on a molar basis.
One exception was the MCF-7 line which is moderately sensitive to temozolomide and has a relatively high ATase activity. Cell lines pretreated with a non-toxic dose of BG were up.to 3.5 fold more and 6 fold more sensitive to temozolomide and CCNU respectively.
The control XP cells (xeroderma pigmentosa cells transfected with pZipneoSV(X)l (Fan et al., Nucleic Acids Res., 18:5723, 1990), which express barely detectable levels of ATase, are 4-5 fold more sensitive to temozolomide or the CCNU-related agent mitozolomide than the human ATase cDNA-transfected cells (see Table In a colony forming assay for the cytotoxicity of temozolomide (see Figure BG pretreatment showed a similar degree of potentiation for the human ATasetransfected XP cells as for the tumour cells, but had no measurable effect on the control XP cells, which do not express ATase. Although BG depleted the ATase activity in the former cells (see below), they remained more resistant to temozolomide than the control pZip transfected fibroblasts.
The repeated dosing schedule data are given below in Table II.
a *o0o 00 0.
WO 94/15615 PCTIGB94/00065 16 TABLE II Repeated Dose Temozolomide Cytotoxicity rIC5. (um)] CELL DAY 1 DAT 2 DAY 3 DAY 4 DAY
LINE
*BG 4BG -BG *BG -BG +BG -BG 4BG *BG 4BG U373 51 18 KAUI 319 196 350 59 383 21 383 7.2 326 MCF-7 319 89 319 51 375 11 Cells were exposed in tissue culture to O6benzylguanine (BG) prior to repeated daily doses of temozolomide.
The repeated dosing schedule showed dramatic potentiation of temozolomide toxicity by BG in MAWI and MCF-7 cells (see also Figure 3).
After treatment with five 24 hour doses, the MAWI cell line was over 300 fold more sensitive to temozolomide when BG was present. Multiple doses of temozolomide, by itself, were not more toxic than a single 24 hour dose in either cell line. In a similar experiment on U373 cells, which have a low level of ATase, the presence of BG caused only a 3 fold potentiation, after four 24 hour doses.
Alkvltransferase levels The concentrations of BG used rapidly reduced to an undetectable level the initially high ATase content of MAWI cells. and human ATase cDNA transfected XP fibroblasts. HPLC analysis showed that BG was stable in tissue culture medium for at least 24 hours at 370C.
Temozolomide, following a three-hour incubation, was found to cause a decrease in ATase content of U373, MCF- 7, LOVO and MAWI cell lines. There was a 50% reduction at 50-100 pM for each line (see Figure despite a 3-4 WO 94/15615 PCT/GB94/00065 17 fold difference in the single dose temozolomide cytotoxicity between MCF-7 and the colorectal lines (LOVO and MAWI). A similar reduction was found in the more sensitive U373 line, although the ATase levels were close to the detection limit of the assay.
To eliminate the possibility of differences in temozolomide transport, the cell uptake of the (14C]labelled compound was studied by the most sensitive and resistant cell lines (ZR-75-1 and MAWI, respectively).
The results shown in Figure 5 show that uptake was very rapid at 4°C, being complete within 5 minutes in both cell lines. Similar amounts of drug were found in both cell lines when adjusted for protein concentration.
Rapid uptake at 40C was consistent with passive diffusion of temozolomide previously shown in two lymphoid lines by Bull et al., Biochem. Pharmacol., 16:3215, (1987).
EXAMPLE 2 Oral Formulation mq. per Capsule Temozolomide 100 20 Lactose, USP 213 Microcrystalline Cellulose Sodium lauryl sulfate Corn starch Magnesium stearate 2 Mix together the temozolomide and lactose, microcrystalline cellulose, sodium lauryl sulfate and corn starch. Pass through a No. 80 screen. Add magnesium stearate, mix and encapsulate into the proper size two-piece gelatin capsule.
o WO 94/15615 PTG9IO6 PCT/GR94/00065 18 EXAMPLE 3 Oral Formulation O'-benzylguanine Lactose, USP Microcrystalline Cellulose Sodium lauryl sulfate Corn starch Magnesium stearate mq. per Capsule 100 213 2 Mix together the 06-benzylguanine, lactose, microcrystalline cellulose, sodium lauryl sulfate and corn starch. Pass through a No. 80 screen. Add magnesium stearate, mix and encapsulate into the proper size two-piece gelatin capsule.
Oral Formulation Temozolomide O'-benzylguanine Lactose, USP Microcrystalline Cellulose Sodium lauryl sulfate Corn starch Magnesium stearate mq. per Capsule 100 100 213 2 Mix together the temozolomide and O'-benzylguanine, lactose, microcrystalline cellulose, s6dium lauryl sulfate and corn starch. Pass through a No. 80 screen.
Add magnesium stearate, mix and encapsulate into the proper size two-piece gelatin capsule.
EXAMPLE Intravenous Formulation Temozolomide mI/ml 100 WO 94/15615 PCT/GB94/00065 19 Sodium Bisulfite, USP 3.2 Disodium Edetate, USP 0.1 Water for Injection, q.s. ad 1.0 ml EXAMPLE 6 Intravenous Formulation Temozolomide 100 O'-benzylguanine 100 Sodium Bisulfite, USP 3.2 Disodium Edetate, USP 0.1 Water for Injection, q.s. ad 1.0 ml This invention may be embodied in other forms or carried out in other ways without departing from the spirit or essential characteristics thereof. The present disclosure is therefore to be considered as in all respects illustrative and not restrictive, the scope of the invention being indicated by the appended Claims, and all changes which come within the meaning and range of equivalency are intended to be embraced therein.

Claims (10)

  1. 6. The method of Claim 1 wherein the dosage 2 administered of the ATase inhibitor is 10-800 mg/kg of 3 patient body weight. 1 7. The method of Claim 6 wherein the temozolomide is 2 administered at a rate of 150-300 mgm 2 of body surface 3 area per day. 1 8. The method of Claim 1 wherein said ATase inhibitor 2 and said temozolomide are administered in divided doses 3 on consecutive days. 21
  2. 9. The method of claim 1 wherein said ATase inhibitor is administered in a dose of 10-800 mg/kg of patient body weight prior to the administration of the temozolomide, said temozolomide is administered in an amount of 150-300 mgm 2 of body surface area per day, and said ATase inhibitor and said temozolomide are administered in divided doses on consecutive days. The method of claim 9 wherein the ATase inhibitor is 0 -benzylguanine.
  3. 11. The method of claim 10 wherein the total dose of temozolomide is divided into at least four individual doses which are administered on at least four consecutive days.
  4. 12. The method of claim 11 wherein said ATase inhibitor is administered two to eight hours prior to the administration of said temozolomide.
  5. 13. A method according to claim 1 wherein the human cancer cells are breast cancer tumour cells, astrocytoma tumour cells, colorectal tumour cells, melanoma tumour S. 20 cells, mycosis fungoides tumour cells or glioma tumour cells.
  6. 14. A pharmaceutical composition, comprising an eo effective amount of an ATase inhibitor and an effective amount of temozolomide, when used for treating human 25 cancer cells in a patient in need of such treatment.
  7. 15. A pharmaceutical composition comprising an ATase inhibiting amount of an ATase-inhibitor and an effective amount of temozolomide, when used in a method as defined in any one of claims 1 to 13.
  8. 16. Use of temozolomide and an ATase inhibitor for the manufacture of a pharmaceutical composition for the treatment of human cancer cells in a patient.
  9. 17. A kit when used in a method as defined in any one of claims 1 to 13, wherein the kit comprises a dosage form of temozolomide and a separate dosage form of an ATase inhibitor.
  10. 30959.DOC -22- 18. A product comprising temozolomide and an ATase inhibitor as a combined preparation when used for separate or sequential administration to a patient in need of such treatment in repeat dosages on consecutive days in the treatment of human cancer cells. 19. A method for potentiating the toxicity of temozolomide in human cancer cells substantially as described herein with reference to Example 1. A pharmaceutical composition comprising an effective amount of an ATase inhibitor and an effective amount of temozolomide, substantially as described herein with reference to Examples 2 to 6. 21. A method for the manufacture of a pharmaceutical composition comprising temozolomide and an ATase 15 inhibitor, substantially as described herein with *S •reference to Examples 2 to 6. Dated this 15th day of February 2000 CANCER RESEARCH CAMPAIGN TECHNOLOGY LIMITED o By their Patent Attorneys GRIFFITH HACK
AU63711/98A 1993-01-14 1998-04-29 Potentiation of temozolomide in human tumour cells Ceased AU721950B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU63711/98A AU721950B2 (en) 1993-01-14 1998-04-29 Potentiation of temozolomide in human tumour cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US004754 1993-01-14
AU58384/94A AU5838494A (en) 1993-01-14 1994-01-13 Potentiation of temozolomide in human tumour cells
AU63711/98A AU721950B2 (en) 1993-01-14 1998-04-29 Potentiation of temozolomide in human tumour cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU58384/94A Division AU5838494A (en) 1993-01-14 1994-01-13 Potentiation of temozolomide in human tumour cells

Publications (2)

Publication Number Publication Date
AU6371198A AU6371198A (en) 1998-06-18
AU721950B2 true AU721950B2 (en) 2000-07-20

Family

ID=3743696

Family Applications (1)

Application Number Title Priority Date Filing Date
AU63711/98A Ceased AU721950B2 (en) 1993-01-14 1998-04-29 Potentiation of temozolomide in human tumour cells

Country Status (1)

Country Link
AU (1) AU721950B2 (en)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ANTI CANCER DRUGS VOL 3, NO 4, AUG 1992, P 401-405 *

Also Published As

Publication number Publication date
AU6371198A (en) 1998-06-18

Similar Documents

Publication Publication Date Title
CA2153775C (en) Potentiation of temozolomide in human tumour cells
US5731304A (en) Potentiation of temozolomide in human tumour cells
Tishler et al. Taxol sensitizes human astrocytoma cells to radiation
Bible et al. Cytotoxic synergy between flavopiridol (NSC 649890, L86-8275) and various antineoplastic agents: the importance of sequence of administration
Ishikawa et al. Positive correlation between the efficacy of capecitabine and doxifluridine and the ratio of thymidine phosphorylase to dihydropyrimidine dehydrogenase activities in tumors in human cancer xenografts
Houghton et al. Antitumor activity of temozolomide combined with irinotecan is partly independent of O 6-methylguanine-DNA methyltransferase and mismatch repair phenotypes in xenograft models
Leamon et al. Comparative preclinical activity of the folate‐targeted Vinca alkaloid conjugates EC140 and EC145
Mattern et al. Synergistic cell killing by ionizing radiation and topoisomerase I inhibitor topotecan (SK&F 104864)
Wang et al. Synthesis and antitumor activity of a novel series of 6-substituted pyrrolo [2, 3-d] pyrimidine thienoyl antifolate inhibitors of purine biosynthesis with selectivity for high affinity folate receptors and the proton-coupled folate transporter over the reduced folate carrier for cellular entry
US7705049B2 (en) Methods for treating non-melanoma cancers with PABA
Kokkinakis et al. Thresholds of O 6-alkylguanine-DNA alkyltransferase which confer significant resistance of human glial tumor xenografts to treatment with 1, 3-bis (2-chloroethyl)-1-nitrosourea or temozolomide
Wickstroem et al. Preclinical combination studies of an FGFR2 targeted thorium-227 conjugate and the ATR inhibitor BAY 1895344
KR19990036136A (en) Use of 1H-1,2,4-triazole derivatives to inhibit cancer growth
Papadopoulou et al. NLCQ-1 (NSC 709257) Exploiting Hypoxia with a Weak DNA-Intercalating Bioreductive Drug
AU721950B2 (en) Potentiation of temozolomide in human tumour cells
US20090203719A1 (en) Enhancing treatment of mdr cancer with adenosine a3 antagonists
NZ536302A (en) Cancer treatment regime with temozolomide
Chow et al. Induction of apoptosis by 2-chloro-2'deoxyadenosine (2-CdA) alone and in combination with other cytotoxic drugs: synergistic effects on normal and neoplastic lymphocytes by addition of doxorubicin and mitoxantrone
US5877215A (en) Method of treating neoplastic cells with prostaglandin and radiation treatment or prostaglandin and platinum-based anti-tumor agents
US20050119289A1 (en) Enhancing treatment of MDR cancer with adenosine A3 antagonists
Riccardi et al. Influence of tetrahydrouridine on the pharmacokinetics of intrathecally administered 1-β-d-arabinofuranosylcytosine
Taft et al. On 01910. Na
O’Dwyer et al. Pharmacology and clinical investigation of SR-2508 (etanidazole)
Imran Methotrexate drug plug and play role in Human body
Resta et al. A phase I study of OSI-461 in combination with mitoxantrone in patients with advanced solid tumors potentially responsive to mitoxantrone

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
SREP Specification republished