WO2003037314A2 - Medical compositions for intravesical treatment of bladder cancer - Google Patents

Medical compositions for intravesical treatment of bladder cancer Download PDF

Info

Publication number
WO2003037314A2
WO2003037314A2 PCT/US2002/035191 US0235191W WO03037314A2 WO 2003037314 A2 WO2003037314 A2 WO 2003037314A2 US 0235191 W US0235191 W US 0235191W WO 03037314 A2 WO03037314 A2 WO 03037314A2
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
cancer
activity
bladder
approximately
Prior art date
Application number
PCT/US2002/035191
Other languages
French (fr)
Other versions
WO2003037314A3 (en
Inventor
Bastiaan Nuijen
Ernie Pfadenhauer
Jos F. Beijen
Original Assignee
Spectrum Pharmaceuticals, Inc.
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 Spectrum Pharmaceuticals, Inc. filed Critical Spectrum Pharmaceuticals, Inc.
Priority to JP2003539658A priority Critical patent/JP4317452B2/en
Priority to EP02786643A priority patent/EP1439835A2/en
Priority to CA2466148A priority patent/CA2466148C/en
Priority to AU2002350115A priority patent/AU2002350115A1/en
Publication of WO2003037314A2 publication Critical patent/WO2003037314A2/en
Publication of WO2003037314A3 publication Critical patent/WO2003037314A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0034Urogenital system, e.g. vagina, uterus, cervix, penis, scrotum, urethra, bladder; Personal lubricants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes

Definitions

  • Bladder cancer accounts for approximately 2% of all malignant cancers and is the fifth and tenth most common cancer in men and women, respectively.
  • the American Cancer Society estimated that 54,500 new cases and 11 ,700 deaths would have occurred in 1997.
  • Superficial bladder cancers (pTa, pT1 and CIS) account for 70-80% of cancers at first presentation. Management of superficial bladder cancer may be achieved by endoscopic surgical resection often followed by a course of adjuvant intravesical chemotherapy or immunotherapy with the aim of both eradicating remaining tumor cells and preventing tumor recurrence (Herr HW (1987) Intravesical therapy — a critical review. Urol Clin N Am 14:399-404).
  • MMC belongs to a class of compounds known as bioreductive drugs (Workman 1994).
  • MMC represents one of the antineoplastic agents used to treat superficial bladder cancers (Maffezzini et al, 1996, Tolley et al, 1996). MMC is activated to a cytotoxic species by cellular reductases although the role of specific reductase enzymes involved in bioreductive activation remains poorly defined and controversial (Cummings et al, 1998a).
  • NQ01 NAD(P)H:Quinone oxidoreductase, EC 1.6.99.2
  • NQ01 a cytosolic flavoprotein which catalyses the two electron reduction of various quinone based compounds using either NADH or NADPH as electron donors
  • E09 (5-aziridinyl- 3-hydroxymethyl-1methyl-2-[1 H-indole-4,7-dione]prop-(3-en-a-ol), is however a much better substrate for NQ01 than MMC (Walton et al, 1991) and a good correlation exists between NQ01 activity and chemosensitivity in vitro under aerobic conditions (Robertson et al, 1994, Fitzsimmons et al, 1996, Smitkamp-Wilms et al, 1994). Under hypoxic conditions however, E09's properties are markedly different with little or no potentiation of E09 toxicity observed in NQ01 rich cells (Plumb and Workman, 1994).
  • Intratumoural administration of E09 to NQ01 rich and deficient tumors produced significant growth delays (although a distinction between damage to the aerobic or hypoxic fraction was not determined) suggesting that if E09 can be delivered to tumors, therapeutic effects may be achieved (Cummings et al, 1998b). While these undesirable characteristics are a serious setback for the treatment of systemic disease, paradoxically they may be advantageous for treating cancers which arise in a third compartment such as superficial bladder cancer.
  • drug delivery is not problematical via the intravesical route and the penetration of E09 into avascular tissue can be increased by maintenance of therapeutically relevant drug concentrations within the bladder (using a one hour instillation period for example). While this method of instilling E09 within the bladder may be useful, there still remains a need for drug delivery vehicles that are capable of delivering an effective amount of E09 within the bladder.
  • the present invention is directed to compositions for treating cancer. More specifically, the compositions of the present invention comprise pharmaceutical products formulated for intravesical instillation to treat bladder cancer.
  • the pharmaceutical products comprise bioredutive alkylating indoloquinone with anti- tumor effects such as, but not limited to, 3-hydroxymethyl-5-aziridinyl-1-1-methyl-2- [1H-indole-4,7-dione]propenol (E09) and a formulation vehicle.
  • the formulation vehicles of the present invention improves the physical characteristics of the solution such as solubility, lyophilization, and ease of reconstitution of the lyophilized solution.
  • the composition of the present invention comprises 3-hydroxymethyl-5-aziridinyl-1-1-methyl-2-[1 H-indole-4,7- dionejpropenol (E09) and a formulation vehicle.
  • the formulation vehicle is a mixture of tert-butanol and water.
  • the formulation vehicle is a mixture of ethanol and water.
  • the formulation vehicle is 2-hydroxypropyl- ⁇ -cyclodextrin.
  • the composition of the present invention comprises E09 and a coating agent.
  • the coating agent allows for better adhesion of the composition to the bladder wall. Consequently, the composition and, in particular, the E09 contacts and may be able to penetrate the avascular tissue that comprises for a time sufficient to treat the bladder cancer.
  • the coating agent is propylene glycol.
  • the coating agent can be selected from the group consisting of hydroxypropylcellulose, carboxymethylcellulose, chitosan hydrochloride, lectin, or polycarbophil.
  • the compositions of the present invention can be delivered to the bladder wall by a liposome.
  • the compositions of the present invention can be delivered to the bladder wall by a microsphere.
  • the compositions of the present invention can be delivered to a patient intravenously.
  • Figure 1 Validation of the polyclonal anti-rat NQ01 antibody for use in immunohistochemical analysis of human NQ01.
  • Panel A Western blot analysis of cell extracts (12.5 p,g protein loaded per lane) for NQ01.
  • Lanes 1-5 represent extracts from DLD-1 (794 ⁇ 121 nmol/min/mg), HT-29 (688 ⁇ 52 nmol/min/mg), H460 (1652 ⁇ 142 nmol/min/mg), MT1 (287 ⁇ 53 nmol/min/mg), and RT112 (30 ⁇ 3 nmol/min/mg) respectively where the values in parenthesis represent NQ01 activity.
  • Lane 6 represents molecular weight markers (ECL protein molecular weight markers, Amersham Pharmacia Biotech, UK).
  • Panel B Western blot analysis using purified human recombinant NQ01. Lanes 1-5 represent protein amounts of 0.25, 0.125, 0.0625, 0.0312 and 0.0156 pmol respectively.
  • Panel C Western blot analysis of cell extracts (25 /,cg protein loaded per lane) derived from H460 cells (lanes 1-2) and BE cells (lanes 3-4).
  • FIG. 1 Immunohistochemical localization of NQ01 in human bladder tumors, normal bladder, urethra and ureter.
  • Tumors (panels A,B and C) were classified as G2 pTa (panel A, [x 200]) and G3 pT2 (panels B [x 100]) and G3 pT4 (panel C [x 200]) which had high to intermediate levels of NQ01 activity as determined by biochemical methods.
  • Panel D (x100) represents a histological section through a macroscopically normal looking section of bladder from a patient who underwent cystectomy for a G3 pT4 tumor ; no tumor was identified in these sections but some inflammatory change was evident.
  • Panels E and F (x200) represent urethra and ureter with no evidence of invasive or in situ carcinoma in these sections. All sections have been stained with NQ01 antibody. Negative staining (without primary antibody) were clear (data not shown).
  • Figure 3 The relationship between NQ01 activity and the response of a panel of cell lines to E09 (panel A) or MMC (panel B) under normal physiological pHe of 7.4 (o) or acidic pHe values of 6.0 ().
  • Figure 4 The relationship between NQ01 activity and the response of a panel of cell lines to E09 (panel A) or MMC (panel B) under normal physiological pHe of 7.4 (o) or acidic pHe values of 6.0 ().
  • the embodiments of the present invention are directed to compositions for treating bladder cancer via intravesical instillation.
  • the composition of the present invention comprises 3-hydroxymethyl-5-aziridinyl-1-1- methyl-2-[1 H-indole-4,7-dione]propenol (E09) and a formulation vehicle.
  • the formulation vehicles of the present invention are solvents that improves the solubility and stability of E09.
  • the formulation vehicles of the present invention can be a mixture of an alcohol and water.
  • E09 dissolves in the formulation vehicles without physical manipulation such as grinding. Because the compositions of the present invention are capable of dissolving greater amounts of E09, additional flexibility with respect to dosage units is achieved.
  • a content of 8.0 mg of E09 per dosage unit is contemplated.
  • instillation doses range from approximately 0.5 mg to approximately 16 mg in a total volume of 40 mL.
  • the formulation vehicles of the present invention are good lyophilization vehicles.
  • the formulation vehicles of the present invention minimizes the time to lyophilize the compositions of the present invention. Accordingly, in one embodiment of the present invention, it is possible to lyophilize the compositions of the present invention in less than approximately 4.5 days.
  • the compositions of the present invention are stable after undergoing lyophilization (see table 4). It is believed that the formulation vehicles of the present invention minimize the crystallization of E09 during the lyophilization process. Consequently, by reducing the amount of crystallization of E09, a smaller volume of fluid is required to reconstitute the compositions of the present invention. As a result, a larger batch size can be achieved due to the reduced reconstitution volumes for the lyophilized composition.
  • the composition of the present invention comprises E09 and a formulation vehicle comprising tert-butanol.
  • the formulation vehicle comprises mixture of ethanol and water.
  • the formulation vehicle is 2- hydroxypropyl- ⁇ -cyclodextrin.
  • the formulation vehicle comprises 40% tert-butanol in water.
  • the amount of tert-butanol may be varied. The tert-butanol solution better dissolves E09 as compared to water.
  • solubility of E09 is at least 9.5 mg/ml whereas the solubility of E09 is approximately 0.2 mg/ml in water. Consequently, a smaller volume of the tert-butanol is required to dissolve a given amount of E09. Additionally, a greater amount of E09 may be dissolved in a given solution. That is, the compositions of the present invention will have a higher concentration of E09 as compared to a solution where E09 is dissolved in water.
  • the composition comprises, E09, a formulation vehicle, and a bulking agent.
  • lactose can be utilized as the bulking agent.
  • the composition of the present invention can be buffered.
  • the composition is buffered to a pH ranging from approximately 9 to approximately 9.5.
  • the composition can be buffered with any known or developed buffering agents.
  • the compositions of the present invention can either be compounded for intravesical delivery or lyophilized. As those skilled in the art will appreciate, the compositions of the present invention can be lyophilized by those methods known or developed in the art.
  • the lyophilized compositions can be reconstituted by a reconstitution vehicle.
  • the reconstitution vehicle comprises 2% sodium bicarbonate, 0.02% disodium edetate and propylene glycol: water (60:40 V/V).
  • This reconstitution vehicle dissolves the lyophilized composition of the present invention and produces a stable solution for administration for up to 24 hours.
  • the reconstitution vehicle of the present invention provides an ampoule having an extractable volume of 5 mL of reconstituted E09 comprising propylene glycol/water/sodium bicarbonate/sodium edetate 60/40/2/0.02% v/v/w/w.
  • the compositions of the present invention also comprises coating agents.
  • the coating agents of the present invention provide better adhesion of the composition to the bladder wall. Consequently, the composition and, in particular, the E09 contacts and may be able to penetrate the avascular tissue that comprises for a time sufficient to treat the bladder cancer.
  • the coating agent is propylene glycol.
  • the coating agent can be selected from the group consisting of hydroxypropylcellulose, carboxymethylcellulose, chitosan hydrochloride, lectin, or polycarbophil.
  • the compositions of the present invention can be delivered to the bladder wall by a liposome.
  • the Iiposomes used are unilamellar or multilamellar and contain at least one cationic phospholipid such as stearylamine, 1 ,2- diacyl-3-trimethylammonium-propane (TAP) or 1 ,2-triacyl-3-dimethylammonium- propane (DAP).
  • the surface Iiposomes may be coated with polyethylene glycol to prolong the circulating half-life of the Iiposomes.
  • neutrally charged Iiposomes such as, but not limited to, phosphatidylcholine and cholesterol can also be used for liposomal entrapment of the compositions of the present invention.
  • the compositions of the present invention can be delivered to the bladder wall by a microsphere such as those known or developed in the art.
  • compositions of the present invention can be delivered to a patient intravenously.
  • the lyophilized composition of the present invention can be reconstituted using the formulation vehicles of the present invention.
  • the reconstituted composition can then be diluted to a desired concentration and delivered to a patient intravenously.
  • the following experiments were conducted to determine the activity of NQ01 in a series of human bladder tumors and normal bladder tissue by both enzymatic and immunohistochemical techniques. Furthermore, the following experiments evaluate strategies for reducing possible system toxicity arising from intravesical therapy based upon the fact that the aerobic activity of E09 against cell lines is enhanced under mild acidic conditions (Phillips et al., 1992). Administration of E09 in an acidic vehicle would result in greater activity within the bladder and any drug absorbed into the blood stream would become relatively inactive due to the rise in extracellular pH. The following experiments also determine the role of NQ01 in the activation of E09 under acidic conditions.
  • NQOI activity Biochemical determination of NQOI activity.
  • Cell cultures in exponential growth were trypsinised, washed twice with Hanks balanced salt solution (HBSS) and sonicated on ice (3 x 30sec bursts at 40% duty cycle and output setting 4 on a Semat 250 cell sonicator).
  • HBSS Hanks balanced salt solution
  • NQ01 activity and protein concentration was determined as described below.
  • Tissues were homogenised (10% w/v homogenate) in sucrose (0.25M) using a Iml tissue homogeniser (Fisher Scientific).
  • Cytosolic fractions were prepared by centrifugation of the homogenate at 18,000 g for 4 min followed by further centrifugation of the supernatant at 110,000 g for 1 h at 4 ° C in a Beckman Optima TL ultracentrifuge. Activity of NQ01 in the supernatant was determined spectrophotometrically (Beckman DU650 spectrophotometer) by measuring the dicumarol sensitive reduction of dichlorophenolindophenol (DCPIP, Sigma Aldrich, UK) at 600 nm (Traver et al, 1992).
  • This assay has been extensively validated for use in measuring NQ01 activity in both tissue and cell homogenates and has been shown to be preferable to other assays for NQ01 activity (Hodnick and Sartorelli, 1997).
  • Each reaction contained NADH (200 IzM), DCPIP (40 /iM, Sigma Aldrich, UK), Dicumarol (20 uM, when required, Sigma Aldrich, UK), cytosolic fraction of tissues (50 p,l per assay) in a final volume of 1 ml Tris HCI buffer (50 mM, pH 7.4) containing bovine serum albumin (0.7 mg ml "1 , Sigma Aldrich, UK).
  • Rates of DCPIP reduction were calculated from the initial linear part of the reaction curve (30s) and results were expressed in terms of nmol DCPIP reduced /min/mg protein using a molar extinction coefficient of 21mNT' cm "1 for DCPIP. Protein concentration was determined using the Bradford assay (Bradford, 1976).
  • the BE cell line has been genotyped for the C609T polymorphic variant of NQOI and is a homozygous mutant (and therefore devoid of NQ01 enzyme activity) with respect to this polymorphism (Traver et al, 1992).
  • Cells were washed in ice cold phosphate buffered saline and lysed by sonication (30 seconds on ice) in Tris HCI (50 mM, pH 7.5) containing 2mM EGTA, 2mM PMSF and 25 Ftg ml "1 leupeptin. Protein concentration was estimated using the Bradford assay (Bradford, 1976) and a total of 12.5 ,ug of protein (in Lamelli sample loading buffer) applied to a 12% SDS-PAGE gel.
  • membranes were blocked in TBS/Tween 20 (0.1%) containing 5% non-fat dry milk for 1 h at room temperature. Membranes were washed in TBS/Tween 20 (0.1 %) prior to the addition of rabbit anti- rat NQ01 antibody (1 :100 dilution) and incubated at room temperature for I h. Membranes were extensively washed in TBS/Tween 20 (0.1%) followed by the addition of anti-rabbit IgG horseraddish peroxidase conjugated secondary antibody (1 :5000 dilution in TBS/Tween 20). Proteins were visualised by ECL based chemiluminescence as described by the manufacturer (Amersham Pharmacia Biotech, Bucks, UK).
  • Sections were then washed and incubated with biotinylated mouse anti rabbit IgG for 30 min prior to immunoperoxidase staining using VECTASTAIN ABC reagents and DAB (Vector Laboratories Ltd, Peterborough, UK). Sections were counterstained with haematoxylin according to standard procedures.
  • H460 human NSCLC cell line was obtained from the American Type Culture Collection (ATCC).
  • ATCC American Type Culture Collection
  • HT-29 human colon carcinoma
  • RT112/83 human bladder carcinoma epithelial
  • EJ138 human bladder carcinoma
  • T24/83 human bladder transitional cell carcinoma
  • All cell lines were maintained as monolayer cultures in RPNII 1640 culture medium supplemented with fetal calf serum (10%), sodium pyruvate (2 mM), L-glutamine (2mM), penicillin/streptomycin (50 lU/ml / 50 jug/ml) and buffered with HEPES (25 mK. All cell culture materials were purchased from Gibco BRL (Paisley, UK). Cells were exposed to MMC or E09 at a range of doses for one hour and chemosensitivity was assessed following a five day recovery period using the MTT assay, details of which have been described elsewhere (Phillips et al, 1992).
  • the pH of the medium used during drug exposure was adjusted using small aliquots of concentrated HCI (40 ,A cone HCI [10.5M] to 20 ml medium gives a pH of 6.0). Calibration curves were conducted over a broad range of pH values in culture medium (pH 3.5 to 11) and the stability of the pH conditions monitored over a one hour incubation period at 37 ° C. At all pH values, no significant changes in the pH of the medium was observed over the one hour drug exposure period (data not presented).
  • HT-29 multicell spheroids were prepared by seeding 5 x 10 5 cells into T25 flasks which had been based coated with agar (1% w/v) and incubated for 24h at 37 C. Immature spheroids were then transferred to a spinner flask (Techne) containing 250m1 of RPMI 1640 growth medium and spheroids were kept in suspension by stirring at 50 rpm. When spheroids reached a diameter of approximately 500 Am, they were harvested for chemosensitivity studies. Multicell spheroids were exposed to a range of E09 concentrations at pHe 6.0 and 7.4 for one hour at 37 ° C.
  • spheroids were washed twice in HBSS prior to dissagregation into single cells using trypsin EDTA. Disaggregated spheroids were then washed in HBSS and then plated into 96 well plates (1 x 10 3 cells per well), and incubated at 37 ° C for four days. Chemosensitivity was assessed using the NM assay as described elsewhere (Phillips et al, 1992).
  • NQ01 The role of NQ01 in the activation of E09 at pHe values of 7.4 and 6.0 was evaluated using the NQ01 inhibitor Flavone Acetic Acid (FAA), details of which are described elsewhere (Phillips, 1999).
  • FAA Flavone Acetic Acid
  • H460 cells H460 cells (NQ01 rich) were plated into 96 well plates at a density of 2 x 10 3 cells per well.
  • Chemosensitivity was determined by the NM assay as described above and results were expressed in terms of IC5 0 values, selectivity ratios (ICs 0 at pHe 7.4 / IC50 at pHe 6.0) and protection ratios (ICSO FAA/E09 combinations / IC50 for E09 alone).
  • NQOI was purified by cybacron blue affinity chromatography, details of which are described elsewhere (Phillips, 1996).
  • the purified protein had a molecular weight of approximately 31 kDa and a specific activity of 139 /Amol DCPIP reduced / min/ mg protein (Phillips, 1996).
  • Reduction of E09 by recombinant human NQ01 was determined at pH 6.0 and 7.4 by measuring the rate of reduction of cytochrome c was measured at 550 nm on a Beckman DU 650 spectrophotometer according to previously published methods (Phillips, 1996). Results were expressed in terms of ,umol cytochrome c reduced / min / mg protein using a molar extinction coefficient of 21.1 mM "1 cm "1 for cytochrome c.
  • Intracellular pH was determined using the fluorescent pH indicator BCECF (2,7-bis-(2-carboxy-ethyl)-5-(and-6) carboxyfluorescein (Molecular Probes, Eugene, USA) according to manufacturers instructions. Confluent flasks of cells were washed with HBSS to remove any traces of serum containing RPMI medium and then incubated with the esterified form of BCECF (BCECF-AM) at a concentration of 2 [tM in HBSS for one hour at 37°C. The non- denaturing detergent Pluronic was added to the probe to aid dispersion.
  • BCECF fluorescent pH indicator
  • BCECF-AM esterified form of BCECF
  • NQ01 activity in tumor and normal bladder specimens The biochemical activity of NQ01 in paired samples of tumor (grade/stage ranging from G2 pTa to G2/G3 T4) and normal bladder mucosa (with three cystectomy specimens) taken from a series of 20 patients is presented in table 1. Within the tumor specimens, a broad range of NQ01 activity existed ranging from 571.4 nmoUmin/mg to undetectable ( ⁇ 0.1 nmol/min/mg). In histologically nonnal bladder mucosa specimens, NQ01 activity ranged from 190.9 to ⁇ 0.1 nmoUmin/mg. In the majority of patients NQ01 activity in the tumor was greater than in the normal bladder mucosa. Tumor grade and stage did not correlate with NQ01 activity (table 1).
  • the activity of E09 was enhanced in both NQ01 rich and deficient cell lines when pHe was reduced to 6.0 and the relationship between NQ01 and chemosensitivity remained good when cells were exposed to E09 under acidic conditions (figure 3). No cell kill was observed in control cultures when the pHe was decreased to 6.0 (in the absence of drug) as determined by the MTT assay.
  • HT-29 multicell spheroids The response of HT-29 multicell spheroids to E09 is presented in figure 4.
  • Spheroids exposed to E09 at pHe 6.0 were significantly more responsive than at pHe 7.4 with IC5o values of 9.89 ⁇ 0.89 and 24.24 ⁇ 3.29 AM respectively.
  • Spheroids were significantly less responsive to E09 than the same cells exposed to E09 as monolayers at both pHe values with ratios of IC50 values for spheroids to monolayers of 202 and 341 at pHe values of 7.4 and 6.0 respectively.
  • the semiquinone produced as a result of one electron reductases would be relatively non " toxic as it would rapidly redox cycle back to the parent compound.
  • Free radical species generated as a result of redox cycling would be detoxified by superoxide dismutase or catalase but under hypoxic conditions, the semiquinone would be relatively stable. If this were the major toxic species, then the activity of E09 against cells with low NQ01 would be potentiated. In NQ01 rich cells however, the major product formed would be the hydroquinone .
  • Aerobic toxicity could be generated as a result of the back oxidation of the hydroquinone to the semiquinone species or the parent quinone (Butler et al, 1996) resulting is free radical generation.
  • hypoxic conditions however the hydroquinone will be more stable and if this is relatively non- toxic, then the activity of E09 against NQ01 cells under hypoxia would not be potentiated.
  • the mechanism of action of E09 under aerobic and hypoxic conditions is complex, the biological data suggest that E09 should target the aerobic fraction of NQ01 rich tumors or the hypoxic fraction of NQOI deficient tumors (Workman, 1994).
  • NQ01 activity in tumor and normal bladder tissues has clearly identified patients whose tumors are either NQ01 rich or NQ01 deficient (table 1). Within the subset of NQ01 rich tumors, enzyme activity is elevated relative to the normal bladder urothelium. Immunohistochemical studies confirm these biochemical measurements with staining confined to tumor cells as opposed to normal stromal cells (fig 2, panels A, B and C). Within normal bladder tissues, NQ01 staining was absent from the urothelial lining of the bladder (fig 2, panel D) and the urethra (fig 2, panel E). Faint staining of the superficial layers of the ureter (fig 2, panel F) was observed although the underlying basal layers of the ureter were negatively stained.
  • hypoxia markers such as pimonidazole (Kennedy et al, 1997) to address this issue and to establish the relationship between NQ01 activity and hypoxia in tumors.
  • E09 is a substrate for other one electron reductases (Maliepand et al, 1995, Saunders et al, 2000) and further studies designed to evaluate whether E09's metabolism by these enzymes is pH dependent needs to be determined.
  • MMC is not metabolised by but is an inhibitor of human kidney NAD(P)H:(quinone acceptor) oxidoreductase. Cancer Chemother Pharmacol 22: 126-130.
  • NAD(P)H quinone oxidoreductase gene expression in human colon carcinoma cells: Characterisation of a mutation which modulates NQ01 activity and mitomycin sensitivity. Cancer Res 52: 797-802. Walton Ml, Smith PJ and Workman P (1991) The role of NAD(P)H:quinone reductase (EC 1.6.99.2, NQ01) in the reductive bioactivation of the novel indoloquinone antitumour agent E09. Cancer Commun 3: 199-206.
  • (C) denotes cystectomy specimens. In all cases, protein levels following preparation of the cytosolic fraction were greater than 0.1 mg/ml. Table 2.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Urology & Nephrology (AREA)
  • Organic Chemistry (AREA)
  • Reproductive Health (AREA)
  • Gynecology & Obstetrics (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Anti-cancer coating compositions comprising 3-hydroxymethyl-5-aziridinyl-1-1-methyl-2-[1H-indole-4,7-dione]propenol (E09) are disclosed. More specifically, the coating compositions comprise EO9 and a formulation vehicle. The formulation vehicle improves the solubility and stability of EO9. Additionally, the coating compositions can include coating agents that provide better adhesion of the coating composition to the bladder wall during intravesical delivery of the coating composition.

Description

MEDICAL COMPOSITIONS FOR INTRAVESICAL TREATMENT OF BLADDER
CANCER
CROSS REFERENCE To RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No. 60/344,446, filed November 1 , 2001 , and whose entire contents are hereby incorporated by reference.
BACKGROUND OF THE INVENTION
Bladder cancer accounts for approximately 2% of all malignant cancers and is the fifth and tenth most common cancer in men and women, respectively. The American Cancer Society estimated that 54,500 new cases and 11 ,700 deaths would have occurred in 1997. Superficial bladder cancers (pTa, pT1 and CIS) account for 70-80% of cancers at first presentation. Management of superficial bladder cancer may be achieved by endoscopic surgical resection often followed by a course of adjuvant intravesical chemotherapy or immunotherapy with the aim of both eradicating remaining tumor cells and preventing tumor recurrence (Herr HW (1987) Intravesical therapy — a critical review. Urol Clin N Am 14:399-404). Both anti-neoplasties (Mitomycin C [MMC], epirubicin and thioTEPA) and immunotherapy (BCG) administered intravesically are effective at reducing tumor recurrence rates although it is unclear whether disease progression to muscle invasive tumors is prevented (Newling D (1990) Intravesical therapy in the management of superficial transitional cell carcinoma of the bladder: the experience of the EORTC GU group, Br J Cancer 61 :497-499; Oosterlink et al. (1993) A prospective European Organization for Research and Treatment of Cancer Genitourinary Group randomized trial comparing transurethral resection followed by a single instillation of epirubicin or water in single stage Ta, T1 papillary carcinoma of the bladder. J Urol 149:749-752). This observation in conjunction with the fact that mortality from bladder cancer is still high underscores the need to develop more effective therapeutic agents (Oosterlink et al. 1993).
One such therapeutic agent is MMC which belongs to a class of compounds known as bioreductive drugs (Workman 1994). MMC represents one of the antineoplastic agents used to treat superficial bladder cancers (Maffezzini et al, 1996, Tolley et al, 1996). MMC is activated to a cytotoxic species by cellular reductases although the role of specific reductase enzymes involved in bioreductive activation remains poorly defined and controversial (Cummings et al, 1998a). This is particularly true for the enzyme NQ01 (NAD(P)H:Quinone oxidoreductase, EC 1.6.99.2) which is a cytosolic flavoprotein which catalyses the two electron reduction of various quinone based compounds using either NADH or NADPH as electron donors (Schlager and Powis, 1988, Siegel et al, 1990). The structurally related compound E09 (5-aziridinyl- 3-hydroxymethyl-1methyl-2-[1 H-indole-4,7-dione]prop-(3-en-a-ol), is however a much better substrate for NQ01 than MMC (Walton et al, 1991) and a good correlation exists between NQ01 activity and chemosensitivity in vitro under aerobic conditions (Robertson et al, 1994, Fitzsimmons et al, 1996, Smitkamp-Wilms et al, 1994). Under hypoxic conditions however, E09's properties are markedly different with little or no potentiation of E09 toxicity observed in NQ01 rich cells (Plumb and Workman, 1994). In NQ01 deficient cell lines however, large hypoxic cytotoxicity ratios have been reported (Workman, 1994). Therefore, E09 has the potential to exploit the aerobic fraction of NQ01 rich tumors or the hypoxic fraction of NQ01 deficient tumors (Workman, 1994).
E09 has been clinically evaluated but despite reports of three partial remissions in phase I clinical trials, no activity was seen against NSCLC, gastric, breast, pancreatic and colon cancers in subsequent phase II trials (Schellens et al, 1994, Dirix et al, 1996). These findings are particularly disappointing in view of the preclinical studies (Hendriks et al, 1993) together with reports that several tumor types have elevated NQ01 levels (Malkinson et al, 1992, Smitkamp-Wilms et al, 1995, Siegel et al, 1998). Several possible explanations have been proposed to explain E09's lack of clinical efficacy (Connors, 1996, Phillips et al, 1998). Recent studies have demonstrated that the failure of E09 in the clinic may not be due to poor pharmacodynamic interactions but may be the result of poor drug delivery to tumors (Phillips et al, 1998). The rapid plasma elimination of E09 (tl/z = 10 min in humans) in conjunction with poor penetration through multicell layers suggests that E09 will not penetrate more than a few microns from a blood vessel within its pharmacokinetic lifespan (Schellens et al, 1994, Phillips et al, 1998). Intratumoural administration of E09 to NQ01 rich and deficient tumors produced significant growth delays (although a distinction between damage to the aerobic or hypoxic fraction was not determined) suggesting that if E09 can be delivered to tumors, therapeutic effects may be achieved (Cummings et al, 1998b). While these undesirable characteristics are a serious setback for the treatment of systemic disease, paradoxically they may be advantageous for treating cancers which arise in a third compartment such as superficial bladder cancer. In this scenario, drug delivery is not problematical via the intravesical route and the penetration of E09 into avascular tissue can be increased by maintenance of therapeutically relevant drug concentrations within the bladder (using a one hour instillation period for example). While this method of instilling E09 within the bladder may be useful, there still remains a need for drug delivery vehicles that are capable of delivering an effective amount of E09 within the bladder.
BRIEF SUMMARY OF THE INVENTION
In a broad aspect, the present invention is directed to compositions for treating cancer. More specifically, the compositions of the present invention comprise pharmaceutical products formulated for intravesical instillation to treat bladder cancer. The pharmaceutical products comprise bioredutive alkylating indoloquinone with anti- tumor effects such as, but not limited to, 3-hydroxymethyl-5-aziridinyl-1-1-methyl-2- [1H-indole-4,7-dione]propenol (E09) and a formulation vehicle. The formulation vehicles of the present invention improves the physical characteristics of the solution such as solubility, lyophilization, and ease of reconstitution of the lyophilized solution.
According to one embodiment of the present invention, the composition of the present invention comprises 3-hydroxymethyl-5-aziridinyl-1-1-methyl-2-[1 H-indole-4,7- dionejpropenol (E09) and a formulation vehicle. According to one embodiment, the formulation vehicle is a mixture of tert-butanol and water. In another embodiment, the formulation vehicle is a mixture of ethanol and water. In yet another embodiment, the formulation vehicle is 2-hydroxypropyl-β-cyclodextrin. These composition embodiments of the present invention can be lyophilized by techniques known or developed in the art. The lyophilized compositions of the present invention are
According to another embodiment of the present invention, the composition of the present invention comprises E09 and a coating agent. The coating agent allows for better adhesion of the composition to the bladder wall. Consequently, the composition and, in particular, the E09 contacts and may be able to penetrate the avascular tissue that comprises for a time sufficient to treat the bladder cancer. In one embodiment of the present invention, the coating agent is propylene glycol. In other exemplary embodiments of the present invention, the coating agent can be selected from the group consisting of hydroxypropylcellulose, carboxymethylcellulose, chitosan hydrochloride, lectin, or polycarbophil. In yet another embodiment of the present invention, the compositions of the present invention can be delivered to the bladder wall by a liposome. In another embodiment, the compositions of the present invention can be delivered to the bladder wall by a microsphere. In another embodiment, the compositions of the present invention can be delivered to a patient intravenously.
BRIEF DESCRIPTION OF DRAWINGS
Figure 1 . Validation of the polyclonal anti-rat NQ01 antibody for use in immunohistochemical analysis of human NQ01. Panel A: Western blot analysis of cell extracts (12.5 p,g protein loaded per lane) for NQ01. Lanes 1-5 represent extracts from DLD-1 (794 ± 121 nmol/min/mg), HT-29 (688 ± 52 nmol/min/mg), H460 (1652 ± 142 nmol/min/mg), MT1 (287 ± 53 nmol/min/mg), and RT112 (30 ± 3 nmol/min/mg) respectively where the values in parenthesis represent NQ01 activity. Lane 6 represents molecular weight markers (ECL protein molecular weight markers, Amersham Pharmacia Biotech, UK). Panel B: Western blot analysis using purified human recombinant NQ01. Lanes 1-5 represent protein amounts of 0.25, 0.125, 0.0625, 0.0312 and 0.0156 pmol respectively. Panel C: Western blot analysis of cell extracts (25 /,cg protein loaded per lane) derived from H460 cells (lanes 1-2) and BE cells (lanes 3-4).
Figure 2. Immunohistochemical localization of NQ01 in human bladder tumors, normal bladder, urethra and ureter. Tumors (panels A,B and C) were classified as G2 pTa (panel A, [x 200]) and G3 pT2 (panels B [x 100]) and G3 pT4 (panel C [x 200]) which had high to intermediate levels of NQ01 activity as determined by biochemical methods. Panel D (x100) represents a histological section through a macroscopically normal looking section of bladder from a patient who underwent cystectomy for a G3 pT4 tumor ; no tumor was identified in these sections but some inflammatory change was evident. Panels E and F (x200) represent urethra and ureter with no evidence of invasive or in situ carcinoma in these sections. All sections have been stained with NQ01 antibody. Negative staining (without primary antibody) were clear (data not shown).
Figure 3. The relationship between NQ01 activity and the response of a panel of cell lines to E09 (panel A) or MMC (panel B) under normal physiological pHe of 7.4 (o) or acidic pHe values of 6.0 (). Regression analysis data (as determined by Sigma Plot graphics) for E09 at pH 7.4 were r = 0.886, slope = -0.52 and at pH 6.0, regression analysis data for E09 was r = 0.804 and slope = -0.51. For MMC, regression analysis at pH 7.4 was r = 0.849, slope = -0.19 and at pH 6.0, r = 0.609, slope = -0.23. Figure 4. Response of HT-29 multicell spheroids following a one hour exposure to E09 under acidic (pHe = 6.0, 0) and physiological (pHe = 7.4, 0) extracellular pH conditions. Values presented are the means of 3 independent experiments ± standard deviation.
DETAILED DESCRIPTION OF THE INVENTION
The embodiments of the present invention are directed to compositions for treating bladder cancer via intravesical instillation. According to one embodiment, the composition of the present invention comprises 3-hydroxymethyl-5-aziridinyl-1-1- methyl-2-[1 H-indole-4,7-dione]propenol (E09) and a formulation vehicle. The formulation vehicles of the present invention are solvents that improves the solubility and stability of E09. In a broad aspect of the present invention, the formulation vehicles of the present invention can be a mixture of an alcohol and water. According to the various embodiments of the present invention, E09 dissolves in the formulation vehicles without physical manipulation such as grinding. Because the compositions of the present invention are capable of dissolving greater amounts of E09, additional flexibility with respect to dosage units is achieved. According to one embodiment, a content of 8.0 mg of E09 per dosage unit is contemplated. In other embodiments, instillation doses range from approximately 0.5 mg to approximately 16 mg in a total volume of 40 mL.
In addition to improving the solubility of E09, the formulation vehicles of the present invention are good lyophilization vehicles. For example, the formulation vehicles of the present invention minimizes the time to lyophilize the compositions of the present invention. Accordingly, in one embodiment of the present invention, it is possible to lyophilize the compositions of the present invention in less than approximately 4.5 days. Furthermore, the compositions of the present invention are stable after undergoing lyophilization (see table 4). It is believed that the formulation vehicles of the present invention minimize the crystallization of E09 during the lyophilization process. Consequently, by reducing the amount of crystallization of E09, a smaller volume of fluid is required to reconstitute the compositions of the present invention. As a result, a larger batch size can be achieved due to the reduced reconstitution volumes for the lyophilized composition.
According to one embodiment, the composition of the present invention comprises E09 and a formulation vehicle comprising tert-butanol. According to another embodiment of the present invention, the formulation vehicle comprises mixture of ethanol and water. In yet another embodiment, the formulation vehicle is 2- hydroxypropyl-β-cyclodextrin. In one exemplary embodiment, the formulation vehicle comprises 40% tert-butanol in water. As those skilled in the art will appreciate, the amount of tert-butanol may be varied. The tert-butanol solution better dissolves E09 as compared to water. By utilizing a tert-butanol formulation vehicle, solubility of E09 is at least 9.5 mg/ml whereas the solubility of E09 is approximately 0.2 mg/ml in water. Consequently, a smaller volume of the tert-butanol is required to dissolve a given amount of E09. Additionally, a greater amount of E09 may be dissolved in a given solution. That is, the compositions of the present invention will have a higher concentration of E09 as compared to a solution where E09 is dissolved in water.
According to another embodiment of the present invention, the composition comprises, E09, a formulation vehicle, and a bulking agent. In one exemplary embodiment, lactose can be utilized as the bulking agent. As those skilled in the art will appreciate, it is contemplated that other bulking agents known or developed in the art may be utilized. According to another exemplary embodiment, the composition of the present invention can be buffered. In one embodiment, the composition is buffered to a pH ranging from approximately 9 to approximately 9.5. The composition can be buffered with any known or developed buffering agents. The compositions of the present invention can either be compounded for intravesical delivery or lyophilized. As those skilled in the art will appreciate, the compositions of the present invention can be lyophilized by those methods known or developed in the art. The lyophilized compositions can be reconstituted by a reconstitution vehicle. According to one exemplary embodiment, the reconstitution vehicle comprises 2% sodium bicarbonate, 0.02% disodium edetate and propylene glycol: water (60:40 V/V). This reconstitution vehicle dissolves the lyophilized composition of the present invention and produces a stable solution for administration for up to 24 hours. Additionally, the reconstitution vehicle of the present invention provides an ampoule having an extractable volume of 5 mL of reconstituted E09 comprising propylene glycol/water/sodium bicarbonate/sodium edetate 60/40/2/0.02% v/v/w/w.
In another aspect of the present invention, the compositions of the present invention also comprises coating agents. The coating agents of the present invention provide better adhesion of the composition to the bladder wall. Consequently, the composition and, in particular, the E09 contacts and may be able to penetrate the avascular tissue that comprises for a time sufficient to treat the bladder cancer. In one embodiment of the present invention, the coating agent is propylene glycol. In other exemplary embodiments of the present invention, the coating agent can be selected from the group consisting of hydroxypropylcellulose, carboxymethylcellulose, chitosan hydrochloride, lectin, or polycarbophil.
In yet another embodiment of the present invention, the compositions of the present invention can be delivered to the bladder wall by a liposome. According to one embodiment of the present invention, the Iiposomes used are unilamellar or multilamellar and contain at least one cationic phospholipid such as stearylamine, 1 ,2- diacyl-3-trimethylammonium-propane (TAP) or 1 ,2-triacyl-3-dimethylammonium- propane (DAP). In another embodiment of the present invention, the surface Iiposomes may be coated with polyethylene glycol to prolong the circulating half-life of the Iiposomes. In yet another embodiment of the present invention, neutrally charged Iiposomes such as, but not limited to, phosphatidylcholine and cholesterol can also be used for liposomal entrapment of the compositions of the present invention. In another embodiment, the compositions of the present invention can be delivered to the bladder wall by a microsphere such as those known or developed in the art.
In yet another embodiment, the compositions of the present invention can be delivered to a patient intravenously. The lyophilized composition of the present invention can be reconstituted using the formulation vehicles of the present invention. The reconstituted composition can then be diluted to a desired concentration and delivered to a patient intravenously.
The following experiments were conducted to determine the activity of NQ01 in a series of human bladder tumors and normal bladder tissue by both enzymatic and immunohistochemical techniques. Furthermore, the following experiments evaluate strategies for reducing possible system toxicity arising from intravesical therapy based upon the fact that the aerobic activity of E09 against cell lines is enhanced under mild acidic conditions (Phillips et al., 1992). Administration of E09 in an acidic vehicle would result in greater activity within the bladder and any drug absorbed into the blood stream would become relatively inactive due to the rise in extracellular pH. The following experiments also determine the role of NQ01 in the activation of E09 under acidic conditions.
Collection of tumor and normal bladder specimens. Ethical approval for tissue collection was obtained from the Local Research Ethical Committee (Bradford NHS Trust) and samples taken from patients following informed consent. A total of 17 paired cold pinch biopsies were taken from bladder tumors and macroscopically normal looking bladder mucosa at cystoscopy, immediately prior to formal transurethral resection of the tumor. Three specimens were taken from patients undergoing cystectomy and tumor and normal samples dissected by pathologists within one hour of surgical removal. Specimens were flash frozen in liquid nitrogen and transported for NQOI enzyme analysis. Further biopsies were taken of the normal bladder mucosa immediately adjacent to the previous biopsy site and sent at the end of the procedure, along with the resected tumor, in formalin for routine histological analysis. In this way bladder tumor and normal bladder urothelium enzymology could be directly correlated with the appropriate tissue histology in each patient. Immunohistochemistry was performed from the subsequently archived wax blocks prepared for histology.
Biochemical determination of NQOI activity. Cell cultures in exponential growth were trypsinised, washed twice with Hanks balanced salt solution (HBSS) and sonicated on ice (3 x 30sec bursts at 40% duty cycle and output setting 4 on a Semat 250 cell sonicator). NQ01 activity and protein concentration was determined as described below. Tissues were homogenised (10% w/v homogenate) in sucrose (0.25M) using a Iml tissue homogeniser (Fisher Scientific). Cytosolic fractions were prepared by centrifugation of the homogenate at 18,000 g for 4 min followed by further centrifugation of the supernatant at 110,000 g for 1 h at 4°C in a Beckman Optima TL ultracentrifuge. Activity of NQ01 in the supernatant was determined spectrophotometrically (Beckman DU650 spectrophotometer) by measuring the dicumarol sensitive reduction of dichlorophenolindophenol (DCPIP, Sigma Aldrich, UK) at 600 nm (Traver et al, 1992). This assay has been extensively validated for use in measuring NQ01 activity in both tissue and cell homogenates and has been shown to be preferable to other assays for NQ01 activity (Hodnick and Sartorelli, 1997). Each reaction contained NADH (200 IzM), DCPIP (40 /iM, Sigma Aldrich, UK), Dicumarol (20 uM, when required, Sigma Aldrich, UK), cytosolic fraction of tissues (50 p,l per assay) in a final volume of 1 ml Tris HCI buffer (50 mM, pH 7.4) containing bovine serum albumin (0.7 mg ml"1, Sigma Aldrich, UK). Rates of DCPIP reduction were calculated from the initial linear part of the reaction curve (30s) and results were expressed in terms of nmol DCPIP reduced /min/mg protein using a molar extinction coefficient of 21mNT' cm "1 for DCPIP. Protein concentration was determined using the Bradford assay (Bradford, 1976).
Immunohistochemistry. Polyclonal antibodies (raised in rabbits) to purified rat NQ01 were a gift from Professor Richard Knox (Enact Pharma Pic). Validation of the antibody for use in immunohistochemistry studies was performed by Western blot analysis using both purified human recombinant NQ01 and cell extracts derived from a panel of cell lines of human origin. These cell lines included H460 (human NSCLC), RT112 (human bladder carcinoma), HT-29 (human colon carcinoma), BE (human colon carcinoma), MT1 (human breast) and DLD-1 (human colon carcinoma). The BE cell line has been genotyped for the C609T polymorphic variant of NQOI and is a homozygous mutant (and therefore devoid of NQ01 enzyme activity) with respect to this polymorphism (Traver et al, 1992). Cells were washed in ice cold phosphate buffered saline and lysed by sonication (30 seconds on ice) in Tris HCI (50 mM, pH 7.5) containing 2mM EGTA, 2mM PMSF and 25 Ftg ml"1 leupeptin. Protein concentration was estimated using the Bradford assay (Bradford, 1976) and a total of 12.5 ,ug of protein (in Lamelli sample loading buffer) applied to a 12% SDS-PAGE gel. Following electrophoretic transfer to nitrocellulose paper, membranes were blocked in TBS/Tween 20 (0.1%) containing 5% non-fat dry milk for 1 h at room temperature. Membranes were washed in TBS/Tween 20 (0.1 %) prior to the addition of rabbit anti- rat NQ01 antibody (1 :100 dilution) and incubated at room temperature for I h. Membranes were extensively washed in TBS/Tween 20 (0.1%) followed by the addition of anti-rabbit IgG horseraddish peroxidase conjugated secondary antibody (1 :5000 dilution in TBS/Tween 20). Proteins were visualised by ECL based chemiluminescence as described by the manufacturer (Amersham Pharmacia Biotech, Bucks, UK).
For immunohistochemical studies, all tissues (both tumor and normal bladder mucosa) were fixed in 10% formalin, processed routinely and embedded in paraffin wax. Two sections of each tissue block were placed on one slide, one section served as the test and the other as a negative control (no primary antibody). A total of 5 sections from each sample were stained for NQ01 (plus negative controls) and tumor and normal samples from a total of 17 patients were analysed. Sections (5 ,um) were dewaxed, rehydrated and incubated with primary antibody (1 :400 dilution) for 4 hours. Sections were then washed and incubated with biotinylated mouse anti rabbit IgG for 30 min prior to immunoperoxidase staining using VECTASTAIN ABC reagents and DAB (Vector Laboratories Ltd, Peterborough, UK). Sections were counterstained with haematoxylin according to standard procedures.
Cell culture and chemosensitivity studies. E09 was a gift from NDDO Oncology, Amsterdam and MMC was obtained from the Department of Pharmacy, St Lukes Hospital, Bradford. H460 (human NSCLC) cell line was obtained from the American Type Culture Collection (ATCC). HT-29 (human colon carcinoma), RT112/83 (human bladder carcinoma epithelial), EJ138 (human bladder carcinoma) and T24/83 (human bladder transitional cell carcinoma) cell lines were obtained from the European Collection of Animal Cell Cultures (ECACC). A2780 (human ovarian carcinoma) and BE (human colon carcinoma) cells were gifts from Dr T Ward (Paterson Institute, Manchester, UK). All cell lines were maintained as monolayer cultures in RPNII 1640 culture medium supplemented with fetal calf serum (10%), sodium pyruvate (2 mM), L-glutamine (2mM), penicillin/streptomycin (50 lU/ml / 50 jug/ml) and buffered with HEPES (25 mK. All cell culture materials were purchased from Gibco BRL (Paisley, UK). Cells were exposed to MMC or E09 at a range of doses for one hour and chemosensitivity was assessed following a five day recovery period using the MTT assay, details of which have been described elsewhere (Phillips et al, 1992). The pH of the medium used during drug exposure was adjusted using small aliquots of concentrated HCI (40 ,A cone HCI [10.5M] to 20 ml medium gives a pH of 6.0). Calibration curves were conducted over a broad range of pH values in culture medium (pH 3.5 to 11) and the stability of the pH conditions monitored over a one hour incubation period at 37°C. At all pH values, no significant changes in the pH of the medium was observed over the one hour drug exposure period (data not presented).
HT-29 multicell spheroids were prepared by seeding 5 x 105 cells into T25 flasks which had been based coated with agar (1% w/v) and incubated for 24h at 37 C. Immature spheroids were then transferred to a spinner flask (Techne) containing 250m1 of RPMI 1640 growth medium and spheroids were kept in suspension by stirring at 50 rpm. When spheroids reached a diameter of approximately 500 Am, they were harvested for chemosensitivity studies. Multicell spheroids were exposed to a range of E09 concentrations at pHe 6.0 and 7.4 for one hour at 37°C. Following drug incubation, spheroids were washed twice in HBSS prior to dissagregation into single cells using trypsin EDTA. Disaggregated spheroids were then washed in HBSS and then plated into 96 well plates (1 x 103 cells per well), and incubated at 37°C for four days. Chemosensitivity was assessed using the NM assay as described elsewhere (Phillips et al, 1992).
The role of NQ01 in the activation of E09 at pHe values of 7.4 and 6.0 was evaluated using the NQ01 inhibitor Flavone Acetic Acid (FAA), details of which are described elsewhere (Phillips, 1999). FAA is a competetive inhibitor of NQ01 with respect to NADH and at a final concentration of 2 mM, inhibition of NQ01 is > 95% whereas the activity of cytochrome P450 reductase and cytochrome b5 reductase is not substantially altered (<5% inhibition). Briefly, H460 cells (NQ01 rich) were plated into 96 well plates at a density of 2 x 103 cells per well. Following an overnight incubation at 37°C, medium was replaced with fresh medium (pH 7.4) containing a non-toxic concentration of FAA (2mM) and incubated for one hour at 37°C. Medium was then replaced with fresh medium containing E09 (range of drug concentrations) and FAA (2mM) at either pHe 7.4 or 6.0. Following a further one hour incubation at 37°C, cells were washed twice with HBSS and incubated at 37°C in growth medium for five days. Chemosensitivity was determined by the NM assay as described above and results were expressed in terms of IC50 values, selectivity ratios (ICs0 at pHe 7.4 / IC50 at pHe 6.0) and protection ratios (ICSO FAA/E09 combinations / IC50 for E09 alone).
Substrate specificity. The influence of acidic pHe on substrate specificity for purified human NQ01 was determined as described previously (Phillips 1996, Walton et al, 1991). NQ01 mediated reduction of the quinone to the hydroquinone species is difficult to detect by conventional assays thereby necessitating the use of a reporter signal generating step. In this assay, the hydroquinone acts as an intermediate electron acceptor which subsequently reduces cytochrome c which can readily be detected spectrophotometrically. Recombinant human NQ01 was derived from E.coli transformed with the pKK233-2 expression plasimd containing the full length cDNA sequence for human NQ01 isolated from the (Beall et al, 1994). Following IPTG induction, NQOI was purified by cybacron blue affinity chromatography, details of which are described elsewhere (Phillips, 1996). The purified protein had a molecular weight of approximately 31 kDa and a specific activity of 139 /Amol DCPIP reduced / min/ mg protein (Phillips, 1996). Reduction of E09 by recombinant human NQ01 was determined at pH 6.0 and 7.4 by measuring the rate of reduction of cytochrome c was measured at 550 nm on a Beckman DU 650 spectrophotometer according to previously published methods (Phillips, 1996). Results were expressed in terms of ,umol cytochrome c reduced / min / mg protein using a molar extinction coefficient of 21.1 mM"1 cm"1 for cytochrome c.
Measurement of intracellular pH. Intracellular pH was determined using the fluorescent pH indicator BCECF (2,7-bis-(2-carboxy-ethyl)-5-(and-6) carboxyfluorescein (Molecular Probes, Eugene, USA) according to manufacturers instructions. Confluent flasks of cells were washed with HBSS to remove any traces of serum containing RPMI medium and then incubated with the esterified form of BCECF (BCECF-AM) at a concentration of 2 [tM in HBSS for one hour at 37°C. The non- denaturing detergent Pluronic was added to the probe to aid dispersion. Cells were then washed to remove all traces of BCECF-AM and then trypsinized before being suspended in serum-free / phenol red-free RPM1 medium (Gibco BRL, Paisley, UK) at a concentration of 106 cells per ml at pH 6 for one hour. Flourescence measurement was determined in a Perkin-Elmer fluorescence spectrophotometer in UV grade disposable 4ml cuvettes (Fischer Scientific) with excitation wavelengths 500nm and 450nm (excitation bandpass slit of lOnm) and emission wavelength fixed at 530nm (emission bandpass slit of 2.5nm). These were determined to be optimal settings for the machine and system under study. An in-situ calibration was performed for every pHi determination with a range of six pH's from 4 to 9 using the ionophore nigericin at a concentration of 22.8 p,M to equilibrate pHe with pHi . Calculation of the ratio of fluorescence at 500nm / 450nm was calculated after subtraction of background fluorescence from blanks at each pH (serum free, phenol red free RPMI without cells).
Activity of NQ01 in tumor and normal bladder specimens. The biochemical activity of NQ01 in paired samples of tumor (grade/stage ranging from G2 pTa to G2/G3 T4) and normal bladder mucosa (with three cystectomy specimens) taken from a series of 20 patients is presented in table 1. Within the tumor specimens, a broad range of NQ01 activity existed ranging from 571.4 nmoUmin/mg to undetectable (< 0.1 nmol/min/mg). In histologically nonnal bladder mucosa specimens, NQ01 activity ranged from 190.9 to < 0.1 nmoUmin/mg. In the majority of patients NQ01 activity in the tumor was greater than in the normal bladder mucosa. Tumor grade and stage did not correlate with NQ01 activity (table 1).
Validation of NQ01 antibody and inununohistochemical localization of NQ01. Western blot analysis demonstrates that polyclonal anti rat NQ01 antibody cross reacts with human NQ01 (figure 1) with a single band at approximately 31 kDa observed for both cell extracts and purified human NQ01. Titration of purified NQO 1 results in a decrease in band intensity (figure 1 B) and in cell extracts, band intensity was qualitatively consistent with NQ01 enzyme activity (figure 1A). In addition, the antibody does not detect NQ01 in the BE cell line which is devoid of NQ01 activity as a result of the C609T polymorphism (figure 1C). No non-specific bands were observed on Western blots. Immunoperoxidase staining of NQ01 protein in tumor tissue, bladder wall, ureter and urethra are presented in figure 2. Superficial and invasive tumors (pTa- panel A, G3 pT2 - panel B and G3pT4 - panel C) with high to intermediate levels of NQ01 as determined by biochemical assays (patient numbers 1 , 4 and 5 in table 1) clearly stained positive for NQ01. Staining was confined to the cytoplasm of tumor cells with little or no staining of stromal cells (panels B and C). In other tumors with intermediate or low levels of NQOI activity, staining was heterogeneous with pockets of cells containing high levels of NQ01 protein (data not shown). Normal bladder wall sections were obtained from a patient who underwent cystectomy (G3pT4 bladder tumor), ureter and urethra were obtained from another patient who underwent cystectomy (G3 pT3a bladder tumor). In the bladder wall, no NQOI staining was observed in the urothelium (panel D) although slight staining was present in smooth muscle layers. The urethra (panel E) was negative although cells on the luminal surface of the ureter were positively stained (panel F). The basal layers of the ureter lining were however negatively stained (panel F). No evidence of invasive malignancy or in situ carcinoma were observed in the ureter and urethra or in the section of bladder wall presented (panel D). In 16 other normal bladder biopsy and cystectomy specimens, no positive staining of the urothelium was observed (data not shown).
Influence of pH on substrate specificity and chemosensitivity. The ability of E09 to serve as a substrate for NQ01 was not influenced by pH with specific activities of 21.10 ± 2.3 and 21.30 ± 1.5 pmol cytochrome c reduced/min/mg protein at pH 7.4 and 6.0 respectively. The response of a panel of cell lines with a range of NQ01 activity (<1.0 to 1 ,898 ± 276 nmol/min/mg) to E09 and MMC at pHe values of 7.4 and 6.0 is presented in table 2 and figure 2. At pHe = 7.4, a good correlation existed between NQ01 activity and chemosensitivity to E09 (figure 3). In the case of MMC (table 2, figure 3), a relationship between NQ01 and chemosensitivity was apparent (at pHe 7.4) although this relationship was not as prominent as shown by E09 with a narrow range of icso values (range 0.9 to 7.0 ttM) observed in cell lines which cover a broad range of NQ01 activity (ranging from <1.0 to 1 ,898 nmol/min/mg). Both MMC and E09 are preferentially more toxic to cells at pHe values of 6.0 although much greater potentiation of E09 activity is seen with SR values (SR = selectivity ratio defined as IC5o pHe 7.4 / icso pHe 6.0) ranging from 3.92 to 17.21 for E09 compared with 1.02 to 4.50 for MMC (table 2). The activity of E09 was enhanced in both NQ01 rich and deficient cell lines when pHe was reduced to 6.0 and the relationship between NQ01 and chemosensitivity remained good when cells were exposed to E09 under acidic conditions (figure 3). No cell kill was observed in control cultures when the pHe was decreased to 6.0 (in the absence of drug) as determined by the MTT assay. The response of H460 cells to E09 at pHe values of 7.4 and 6.0 in the presence and absence of FAA (2mM) is presented in table 3. At both pHe values, the response of H460 cells to E09 was reduced in the presence of FAA. Protection ratios defined as the IC50 for E09 plus FAA divided by the IC50 value for E09 alone were similar for cells under acidic and physiological pHe values (14.63 and 13.95 respectively, table 3). Selectivity ratios defined as the IC50 at pHe 7.4 divided by the IC50 at pHe 6.0 in the presence and absence of FAA were also similar with SR values of 6.31 and 6.02 for E09 alone and E09 plus FAA respectively (table 3). The response of HT-29 multicell spheroids to E09 is presented in figure 4. Spheroids exposed to E09 at pHe 6.0 were significantly more responsive than at pHe 7.4 with IC5o values of 9.89 ± 0.89 and 24.24 ± 3.29 AM respectively. Spheroids were significantly less responsive to E09 than the same cells exposed to E09 as monolayers at both pHe values with ratios of IC50 values for spheroids to monolayers of 202 and 341 at pHe values of 7.4 and 6.0 respectively.
Influence of acidic pHe conditions on pHi. PM values following a one hour incubation at pHe 6.0 were 6.44 ± 0.04, 6.51 ± 0.02 and 6.42 ± 0.05 in A549, RT112/83 and A2780 cells respectively. Addition of the ionophore nigericin (after a one hour incubation at pHe 6.0) resulted in the equilibration of pHe and p11i.
In terms of bioreductive drug development, two of the critical factors which will ultimately determine selectivity are the enzymology of tumors and the presence of hypoxia (Workman, 1994). As outlined in the introduction, the presence or absence of NQ01 is central to the design of appropriate E09 based therapeutic strategies aimed at targeting either the aerobic (NQ01 rich cells) or hypoxic fraction (NQ01 deficient tumors) of tumors. Workman (1994) has outlined a proposed mechanism for the different properties of E09 under aerobic and hypoxic conditions based on the hypothesis that it is the semiquinone (product of one electron reduction) rather than the hydroquinone which is responsible for toxicity. In NQOI deficient cells, the semiquinone produced as a result of one electron reductases would be relatively non" toxic as it would rapidly redox cycle back to the parent compound. Free radical species generated as a result of redox cycling would be detoxified by superoxide dismutase or catalase but under hypoxic conditions, the semiquinone would be relatively stable. If this were the major toxic species, then the activity of E09 against cells with low NQ01 would be potentiated. In NQ01 rich cells however, the major product formed would be the hydroquinone . Aerobic toxicity could be generated as a result of the back oxidation of the hydroquinone to the semiquinone species or the parent quinone (Butler et al, 1996) resulting is free radical generation. Under hypoxic conditions however the hydroquinone will be more stable and if this is relatively non- toxic, then the activity of E09 against NQ01 cells under hypoxia would not be potentiated. Whilst the mechanism of action of E09 under aerobic and hypoxic conditions is complex, the biological data suggest that E09 should target the aerobic fraction of NQ01 rich tumors or the hypoxic fraction of NQOI deficient tumors (Workman, 1994).
Analysis of NQ01 activity in tumor and normal bladder tissues has clearly identified patients whose tumors are either NQ01 rich or NQ01 deficient (table 1). Within the subset of NQ01 rich tumors, enzyme activity is elevated relative to the normal bladder urothelium. Immunohistochemical studies confirm these biochemical measurements with staining confined to tumor cells as opposed to normal stromal cells (fig 2, panels A, B and C). Within normal bladder tissues, NQ01 staining was absent from the urothelial lining of the bladder (fig 2, panel D) and the urethra (fig 2, panel E). Faint staining of the superficial layers of the ureter (fig 2, panel F) was observed although the underlying basal layers of the ureter were negatively stained. Similarly, faint staining of the smooth muscle layers of the bladder, ureter and urethra were also observed (data not shown). These studies suggest that a proportion of patients with bladder tumors (at various grades and stages of the disease) exhibit a significant differential in terms of NQ01 activity which could potentially be exploited by E09 based therapies directed against the aerobic fraction of tumor cells. With regards to the ability of E09 to selectively kill hypoxic NQ01 deficient cells, a subset of patients also exist whose tumors are devoid of NQ01 activity (table 1). It is not known whether or not bladder tumors contain regions of low oxygen tension and further studies are required using hypoxia markers such as pimonidazole (Kennedy et al, 1997) to address this issue and to establish the relationship between NQ01 activity and hypoxia in tumors.
Whilst biochemical and immunohistochemical studies demonstrate that a subset of patients exist which have the appropriate tumor enzymology to activate E09 (under aerobic conditions), intravesical chemotherapy can result in systemic toxicity due to the drug entering the blood supply. This study has also evaluated a potential strategy for minimizing any risk of systemic toxicity based upon the hypothesis that administration of E09 in an acidic vehicle would enhance the potency of E09 (Phillips et al, 1992) within the bladder and that any drug reaching the blood stream would become relatively inactive due to a rise in pHe. Selectivity for aerobic cells would still be determined by NQ01 activity and therefore it is essential to determine the role that NQ01 plays in the activation of E09 under acidic pHe conditions. In a panel of cell lines with a broad spectrum of NQ01 activity, reducing the pHe to 6.0 enhances the potency of E09 under aerobic conditions in all cases (with SR values ranging from 3.92 to 17.21 , table 2). In the case of MMC, potency is also enhanced at low pHe values although the magnitude of the pH dependent increase in toxicity is reduced (SR values ranging from 1.02 to 4.50, table 2) compared with E09. With respect to MMC, one explanation for increased activity under acidic conditions has been attributed to the fact that MMC becomes a substrate for NQ01 under acidic conditions (Pan et al, 1993, Siegel et al, 1993). This is not the case with E09 as rates of reduction of E09 by purified human NQ01 are not influenced by pH (21.10 ± 2.30 and 21.30 ± 1.50 limol cytochrome c reduced / min / mg protein at pH 7.4 and 6.0 respectively). Recent studies have demonstrated that the activity of E09 is enhanced under acidic conditions (pHe = 6.5) but only when the intracellular pH is reduced (plli = 6.5) by co-incubation with nigericin (Kuin et al, 1999). The results of this study are in agreement with this finding as pHi becomes acidic (pHi values range from 6.42 ± 0.05 to 6.51 ± 0.02 depending on the cell line) when cells are cultured under pHe 6.0 conditions.
In the panel of cell lines used in this study, a good correlation exists between NQ01 activity and chemosensitivity at both pHe values of 7.4 and 6.0 (figure 3). A strong relationship between NQ01 activity and response under aerobic conditions (at pHe 7.4) has been established previously by several groups (Robertson et al, 1994, Fitzsimmons et al, 1996, Smitkamp-Wilms et al, 1994) and there is clear evidence that NQ01 plays a central role in the mechanism of action of E09 under aerobic conditions (Workman, 1994). The good correlation between NQ01 activity and response at pHe 6.0, in conjunction with the fact that E09 is still a good substrate for NQ01 at pH 6.0, suggests that NQ01 plays a significant role in E09's mechanism of action at acidic pHe values under aerobic conditions. It is of interest to note however that the activity of E09 against BE cells (which are devoid of NQ01 activity as a result of the C609T polymorphism, Traver et al, 1992) is also enhanced under acidic pHe conditions (table 2). This suggests that there is a NQ01 independent mechanism for the increased activity of E09 under acidic conditions. This is confirmed by the use of the NQ01 inhibitor FAA where the 'protection ratios' (defined as the ratio of IC5o values for E09 plus FAA divided by the ICSo values for E09) are similar at both pHe 7.4 and 6.0 (13.95 and 14.63 respectively, table 3). If NQ01 played a central role in the activation of E09 at pHe 6.0, then the protection ratio at pHe 6.0 would be significantly greater than the protection ratio at pHe 7.4. The mechanism behind the NQ01 independent activation of E09 is unclear although it is a well known fact that the reactivity of aziridine ring structures is enhanced by protonation resulting in ring opening to the aziridinium ion which is a potent alkylating species (Mossoba et al, 1985, Gutierrez, 1989). Alternatively, E09 is a substrate for other one electron reductases (Maliepaard et al, 1995, Saunders et al, 2000) and further studies designed to evaluate whether E09's metabolism by these enzymes is pH dependent needs to be determined. The potency of E09 can be enhanced further by reducing pHe below 6.0 (Phillips et al, 1992) but these conditions are unlikely to provide significant clinical benefits as E09 becomes progressively more unstable when pH is reduced to 5.5 (t'/s = 37 min). From a pharmacological standpoint, administration of E09 in a vehicle at pH 6.0 would appear desirable. Not only would this result in significant enhancement of E09 activity but also the stability of E09 would be sufficient (tlh = 2.5 h) to maintain drug exposure parameters at a therapeutic level.
With regards to the activity of E09 against three dimensional culture models in vitro, this study has demonstrated that reducing the pHe to 6.0 enhances the potency of E09 against multicell spheroids although the magnitude of this effect is reduced compared with monolayer cultures (figure 4). It is not known whether or not reduction in pHe results in greater cell kill throughout the spheroid or if it is confined to the surface of the spheroid exposed to medium. In comparison with MMC, previous studies using histocultures exposed to MMC demonstrated that no difference in toxicity exists between physiological and acidic pHe conditions (Yen et al, 1996). The pH dependent increase in E09 toxicity against spheroids suggests that manipulation of pHe may not only be of use in treating a multilayered solid bladder tumor but may offer an advantage over MMC. It should however be stated that multicell spheroids are significantly less responsive to E09 than mono] ayers, presumably because of the poor penetration properties of E09 through avascular tissue (Phillips et al, 1998). E09 can nevertheless kill >90% of cells in spheroids (figure 4) suggesting that a higher doses at least, the penetration of E09 is sufficient to eradicate cells which reside some distance away from the surface of the spheroid.
In conclusion, the results of this study have demonstrated that within a population of patients with bladder tumors at various stages and grades of the disease, there exists a great heterogeneity regarding the expression of NQ01. The majority of patients have tumors possessing elevated levels of NQO 1 while a small subset of patients appear to be devoid of NQ01 activity. The heterogeneous nature of NQ01 activity described here is consistent with several other studies in various tumor types (Malkinson et al, 1992, Smitkamp-Wilms et al, 1995, Siegel et al, 1998). These findings reinforce the view that 'enzyme profiling' of individual patients could be valuable prior to therapeutic intervention with bioreductive drugs (Workman, 1994). This is to our knowledge the first study to characterize NQ01 activity and cellular localization in bladder tumors and provide strong evidence to support the evaluation of E09 against superficial and locally invasive bladder tumors. This study has clearly demonstrated that under aerobic conditions, E09 is much more potent under acid conditions (pH6.0) than at physiological pH (pH7.4). The mechanism for this increased E09 potency appears to be NQ01 independent and whilst this will not improve (or reduce) selectivity, it may prove beneficial in terms of reducing the therapeutically effective dose of E09. Dose reduction in conjunction with the fact that a reduction in the potency of E09 due to the increased pHe in the blood stream suggests that systemic toxicity arising from the intravesical administration of E09 would be low. In addition, this study shows that under physiological conditions the activity of E09 is much lower in tissues with "normal" expression of NQ01 compared to "high" NQ01 expressing tissues (i.e. the tumors). The results of this study provide strong evidence in support of the proposal that intravesical administration of E09 may have activity against bladder tumors.
REFERENCES.
Butler J, Spanswick VJ and Cummings J (1996) The autooxidation of reduced forms of E09. Free Rad Res 25: 141-148.
Bradford MM (1976) A rapid and sensitive method for the quantification of microgram quantities of protein utilising the principle of protein-dye binding. Anal Biochem 72: 248-254.
Connors TA (1996) Bioreductive agents, hypoxic cells and therapy. EurJ Cancer 32A: 1833-1834.
Cummings J, Spanswick VJ, Tomaz M and Smyth JF (1998a) Enzymology of MMC metabolic activation in tumor tissue. Implications for enzyme directed bioreductive drug development. Biochem Pharmacol 56: 405-414.
Cummings J, Spanswick VJ, Gardiner J, Ritchie A and Smyth, IF (1998b) Pharmacological and biochemical determinants of the antitumour activity of the indoloquinone E09. Biochem Pharmacol 55: 253-260.
Dirix LY, Tonnesen F, Cassidy J, Epelbaum R, Huinink WWT, Pavlidis N, Sorio R, Gamucci T, Wolff I, Tevelde A, Lan J, and Verweij J (1996) E09 phase II study in advanced breast, gastric, pancreatic and colorectal carcinoma by the early clinical studies group. EurJ Cancer 32A: 2019-2022.
Fitzsimmons S A, Workman P, Grever M, Paull K, Camalier R and Lewis AD (1996) Reductase enzyme expression across the National Cancer Institute tumor cell line panel: Correlation with sensitivity to MMC and E09. J Natl Cancer Inst 88: 259- 269 Gutierrez PL (1989) Mechanism of bioreductive alkylation. The example of diazaquinone (AZQ. Free Radical Bio Med 6: 405-445.
Hendriks HR, Piazo PE, Berger DP, Kooistra KL, Bibby MC, Boven E, Dreef-Van Der Meulen HC, Henrar-REC, Fiebig HH, Double JA, Hornstra HW, Pinedo HM, Workman P and Swartsmann G (1993) E09: A novel bioreductive alkylating indoloquinone with preferential solid tumor activity and lack of bone marrow toxicity in preclinical models. EurJ Cancer 29A: 897-906.
Herr HW (1987) Intravesical therapy - a critical review. Urology Clinics of N America 14: 399-404.
Hodnick WF and Sartorelli AC (1997) Measurement of dicumarol sensitive NADPH: (menadione cytochrome c) oxidoreductase activity results in an artifactual assay of DT-diaphorase in cell sonicates. Anal Biochem 252: 165-168.
Kennedy AS, Raleigh JA, Perez GM, Calkins DP, Thrall DE, Novotny DB, Varia MA (1997) Proliferation and hypoxia in human squamous cell carcinoma of the cervix: first report of combined immunohistochemical assays. Int J Radiat Oncol Biol Phys 37: 897-905.
Kuin A, Alders M, Lamfers M, Van Zuidam DJ, Essers M, Beijnen JH and Smets LA (1999) Potentiation of anti-cancer activity at low intratumoural pH induced by the mitochondrial inhibitor m-iodobenzylguanidine (MIBG) and its analogue benzylguanidine (BG). BrJ Cancer 79: 793-801.
Maffezzini M, Simonata A, Zanon M, Raber M and Carmig ani G (1996) Up- front chemotherapy for low stage low grade recurrent bladder cancer. J Urol 155: 91- 93. Malkinson AM, Siegel D, Forrest GL, Gazdar AF, Oie HK, Chan DC, Bunn PA, Mabry M, Dykes DJ, Harrison SD and Ross D (1992) Elevated NQ01 activity and messenger RNA content in human non small cell lung carcinoma - Relationship to the response of lung tumor xenografts to MMC. Cancer Res 52: 4752-4757.
Maliepaard M, Wolf A, Groot SE, De Mol NJ and Janssen LHM (1995) Indoloquinone E09: DNA interstrand cross linking upon reduction by DT-diaphorase or xanthine oxidase. BrJ Cancer 71: 836-839. Mossoba MM, Alizadeh M and Gutierrez PL (1985) Mechanism for the reductive activation of diazaquinone. J Pharm Sci 74: 1249-1254.
Newling D (1990) Intravesical therapy in the management of superficial transitional cell carcinoma of the bladder: the experience of the EORTC GU group. Br J Cancer61 : 497-499.
Oosterlink W, Kurth KH, Schrδder F, Bultinck J, Hammond B, Sylvester R, and members of the European Organisation for Research and Treatment of Cancer Genitourinary Group (1993) A prospective European Organisation for Research and Treatment of Cancer Genitourinary Group randomised trial comparing transurethral resection followed by a single instillation of epirubicin or water in single stage Ta, T 1 papillary carcinoma of the bladder. J Urol 149: 749-752.
Pan SS, Yu F and Hipsher C (1993) Enzymatic and pH modulation of MMC induced DNA damage in MMC resistant HCT 116 human colon cancer cells. Mol Pharmacol 43: 870-877.
Phillips RM, Hulbert PB, Bibby MC, Sleigh NR, and Double JA (1992) In vitro activity of the novel indoloquinone EO-9 and the influence of pH on cytotoxicity. BrJ Cancer 65: 359-364.
Phillips RM (1996) Bioreductive activation of a series of analogues of 5- aziridinyl-3hydroxymethyl-l-methyl-2-[1 H-indolo-4,7-dione] prop-(3-en-a-ol (E09) by human NQ01. Biochem Pharmacol 52: 1711-1718.
Phillips RM, Loadman PM and Cronin BP (1998) Evaluation of a novel in vitro assay for assessing drug penetration into avascular regions of tumors. BrJ Cancer 77: 2112-2119.
Phillips RM (1999) Inhibition of DT-diaphorase (NAD(P)H:quinone oxidoreductase, EC 1.6.99.2) by 5,6-dimethylxanthenone-4-acetic acid (DNIXAA) and flavone-δacetic acid (FAA): Implications for bioreductive drug development. Biochem Pharmacol 58: 303-310.
Plumb JA and Workman P (1994) Unusually marked hypoxic sensitisation to indoloquinone E09 and MMC in a human colon tumor cell line that lacks NQOI activity. Int J Cancer 56: 134-139.
Robertson N, Haigh A, Adams GE and Stratford U (1994) Factors affecting sensitivity to E09 in rodent and human tumor cells in vitro: NQ01 activity and hypoxia. EurJ Cancer 30A: 1013-1019.
Saunders MP, Jaffar M, Patterson AV, Nolan J, Naylor MA, Phillips RM, Harris AL and Stratford H (2000) The relative importance of NADPH:cytochrome c (P450) reductase for determining the sensitivity of human tumor cells to the indoloquinone E09 and related analogues lacking functionality at the C-2 and C-3 positions. Biochem Pharmacol 59: 993-996.
Schellens JHM, Planting AST, Van Acker BAC, Loos WJ, De Boer-Dennert M, Van Der Burg MEL, Koier I, Krediet RT, Stoter G and Verweij J (1994) Phase I and pharmacologic study of the novel indoloquinone bioreductive alkylating cytotoxic drug E09. J Natl Cancer Inst 86: 906-912.
Schlager JJ and Powis G (1988) MMC is not metabolised by but is an inhibitor of human kidney NAD(P)H:(quinone acceptor) oxidoreductase. Cancer Chemother Pharmacol 22: 126-130.
Siegel D, Gibson NW, Preusch PC and Ross D (1990) Metabolism of MMC by NQ01 : Role in MMC induced DNA damage and cytotoxicity in human colon carcinoma cells. Cancer Res 50: 7483-7489.
Siegel D, Beall HD, Kasai M, Gibson NW and Ross D (1993) PH dependent inactivation of NQ01 by MMC and porfiromycin. Mol Pharmacol 44: 1128-1134. Siegel D, Franklin WA and Ross D (1998) Immunohistochemical detection of NAD(P)H:Quinone oxidoreductase in human lung and lung tumors. Clin Cancer Res 4: 2065-2070.
Smitkamp-Wilms E, Peters GJ, Pinedo HM, Van Arkotte J and Giaccone G (1994) Chemosensitivity to the indoloquinone E09 is correlated with NQ01 activity and gene expression. Biochem Pharmacol 47: 1325-1332.
Smitskamp-Wilms E, Giaccone G, Pinedo HM, Van Der Laan BFAM and Peters GJ (1995) NQ01 activity in normal and neoplastic human tissues: An indicator of sensitivity to bioreductive agents ?. Br J Cancer 72: 917-921.
Tolley DA, Parmar MKB, Grigor KM, Lallemand G and the Medical Research Council superficial bladder cancer working party (1996) The effect of intravesical
MMC on recurrence of newly diagnosed superficial bladder cancer: A further report with 7 years of followup. J Urol 155: 1233-1238.
Traver RD, Horikoshi T, Dannenberg KD, Stadlbauer THW, Dannenberg PV, Ross D and Gibson NW (1992) NAD(P)H:quinone oxidoreductase gene expression in human colon carcinoma cells: Characterisation of a mutation which modulates NQ01 activity and mitomycin sensitivity. Cancer Res 52: 797-802. Walton Ml, Smith PJ and Workman P (1991) The role of NAD(P)H:quinone reductase (EC 1.6.99.2, NQ01) in the reductive bioactivation of the novel indoloquinone antitumour agent E09. Cancer Commun 3: 199-206.
Workman P (1994) Enzyme directed bioreductive drug development revisited: A commentary on recent progress and future prospects with emphasis on quinone anticancer drugs and quinone metabolising enzymes, particularly NQ01. Oncol Res 6: 461-475.
Yen WC, Schmittgen T and Au JL (1996) Different pH dependency of mitomycin C activity in monolayer and three dimensional cultures. Pharmaceut Res 13: 1887-1891.
Table 1.
Tumor histology reports and NQ01 activity in paired samples of bladder tumor and normal bladder mucosa.
Figure imgf000023_0001
mMale, Female,s Smoker, 'Non-smoker, e Ex-smoker, Intravesical chemotherapy prior to specimen collection, r Radiotherapy prior to specimen collection,' First presentation, P Previous malignancy other than bladder, h No medical history available, ° Possible occupational carcinogen exposure (i.e., dye industry worker).
(C) denotes cystectomy specimens. In all cases, protein levels following preparation of the cytosolic fraction were greater than 0.1 mg/ml. Table 2.
The relationship between NQ01 activity and chemosensitivity to E09 and MMC under physiological and acidic pHe conditions.
Figure imgf000024_0001
All results presented are the mean of 3 independent experiments (SD values omitted in the interests of presentation).
* SR (selectivity ratio) = IC5o at pH 7.4 / IC5o at pH 6.0
Table 3.
Response of H460 cells to E09 in the presence or absence of FAA (2mm) at pHe values of 7.4 and 6.0.
ICso
Drug pHe SR* PR* (nM)
E09 7.4 60.0 ± 8.1
E09 6.0 9.5 ± 2.6 6.31
E09/FAA 7.4 837 ± 45 13.95
E09/FAA 6.0- 139 ± 27 6.02 14.63
* SR = Selectivity Ratio defined as the ratio of ICSo values at pHe =7.4 divided by the
IC50 at pHe = 6.0.
** Plf = Protection ratio defined as the ratio of IC50 values for E09 plus FAA divided by the IC5o values for E09 alone.
All values represent the mean ± standard deviation for three independent experiments. Table 4 Neoquin 8mg/vial lyophilised product
Figure imgf000025_0001
"purity as chromatographic purity n=3

Claims

What is claimed is:
1. An anti-cancer formulation comprising: a buffered solution comprising 3-hydroxymethyl-5-aziridinyl-1-1-methyl- 2-[1H-indole-4,7-dione]propenol and a formulation vehicle.
2. The anti-cancer formulation of claim 1 wherein the anti-cancer formulation is lyophilized.
3. The anti-cancer formulation of claim 1 wherein the buffered solution has a pH ranging from approximately 9 to approximately 9.5.
4. The anti-cancer formulation of claim 1 wherein the formulation vehicle is selected from the group consisting of tert-butanol/water, ethanol/water, and 2- hydroxypropyl-β-cyclodextrin solution.
5. The anti-cancer formulation of claim 1 further comprising a bulking agent.
6. The anti-cancer formulation of claim 5 wherein the bulking agent is lactose.
7. A anti-cancer formulation for treating bladder cancer, the formulation comprising: a buffered, lyophilized solution comprising 3-hydroxymethyl-5-aziridinyl- 1-1-methyl-2-[1 H-indole-4,7-dione]propenol and a formulation vehicle.
8. The anti-cancer formulation of claim 7 wherein the buffered, lyophilized solution has a pH ranging from approximately 9 to approximately 9.5.
9. The anti-cancer formulation of claim 7 wherein the formulation vehicle is selected from the group consisting of tert-butanol/water, ethanol/water, and 2- hydroxypropyl-β-cyclodextrin solution.
10. The anti-cancer formulation of claim 7 further comprising a bulking agent.
11. The anti-cancer formulation of claim 10 wherein the bulking agent is lactose.
12. An anti-cancer formulation comprising: 3-hydroxymethyl-5-aziridinyl-1-1-methyl-2-[1 H-indole-4,7-dione]propenol dissolved in a buffered formulation vehicle, the buffered formulation vehicle having a pH ranging from approximately 9 to approximately 9.5, and wherein the formulation vehicle is selected from the group consisting of tert-butanol/water, ethanol/water, and 2-hydroxypropyl-β-cyclodextrin solution.
13. The anti-cancer formulation of claim 12 further comprising a bulking agent.
14. The anti-cancer formulation of claim 13 wherein the bulking agent is lactose.
15. An anti-cancer formulation comprising: an indolequinone having a pH within a range of approximately 9 and approximately 9.5, a refractive index within a range of approximately 1.393 to approximately 1.406, and a relative density of within a range of approximately 0.94 to approximately 0.95.
16. The anti-cancer formulation of claim 15 wherein the indolequinone is 3- hydroxymethyl-5-aziridinyl-1-1-methyl-2-[1 H-indole-4,7-dione]propenol.
17. The anti-cancer formulation of claim 16 further comprising a coating composition.
18. The anti-cancer formulation of claim 17 wherein the coating composition is selected from the group consisting of propylene glycol, hydroxypropylcellulose, carboxymethylcellulose, chitosan hydrochloride, lectin, and polycarbophil.
PCT/US2002/035191 2001-11-01 2002-11-01 Medical compositions for intravesical treatment of bladder cancer WO2003037314A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2003539658A JP4317452B2 (en) 2001-11-01 2002-11-01 Medical composition for intravesical treatment of bladder cancer
EP02786643A EP1439835A2 (en) 2001-11-01 2002-11-01 Medical compositions for intravesical treatment of bladder cancer
CA2466148A CA2466148C (en) 2001-11-01 2002-11-01 Medical compositions for intravesical treatment of bladder cancer
AU2002350115A AU2002350115A1 (en) 2001-11-01 2002-11-01 Medical compositions for intravesical treatment of bladder cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US34444601P 2001-11-01 2001-11-01
US60/344,446 2001-11-01

Publications (2)

Publication Number Publication Date
WO2003037314A2 true WO2003037314A2 (en) 2003-05-08
WO2003037314A3 WO2003037314A3 (en) 2003-10-16

Family

ID=23350584

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/035191 WO2003037314A2 (en) 2001-11-01 2002-11-01 Medical compositions for intravesical treatment of bladder cancer

Country Status (13)

Country Link
US (6) US6894071B2 (en)
EP (3) EP1439835A2 (en)
JP (1) JP4317452B2 (en)
AT (2) ATE455545T1 (en)
AU (1) AU2002350115A1 (en)
CA (2) CA2466148C (en)
CY (1) CY1111252T1 (en)
DE (2) DE60238798D1 (en)
DK (2) DK1864660T3 (en)
ES (2) ES2384208T3 (en)
HK (1) HK1132181A1 (en)
PT (2) PT2060259E (en)
WO (1) WO2003037314A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006105507A2 (en) * 2005-03-31 2006-10-05 Spectrum Pharmaceuticals, Inc. Indoloquinone tumor radiation sensitization
WO2007092964A1 (en) * 2006-02-09 2007-08-16 Spectrum Pharmaceuticals, Inc. Bladder cancer treatment by using e09 and propylene glycol
US8563592B2 (en) 2001-11-01 2013-10-22 Spectrum Pharmaceuticals, Inc. Bladder cancer treatment and methods
US8648108B2 (en) 2001-11-01 2014-02-11 Spectrum Pharmaceuticals, Inc. Medical compositions for intravesical treatment of bladder cancer
WO2021008520A1 (en) * 2019-07-15 2021-01-21 深圳艾欣达伟医药科技有限公司 Stable ast-3424 injection preparation and preparation method

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8361490B2 (en) 2004-09-16 2013-01-29 Theracoat Ltd. Biocompatible drug delivery apparatus and methods
ATE540996T1 (en) 2005-06-06 2012-01-15 Univ British Columbia POLYMER BASED SERUM ALBUMINE SUBSTITUTE
WO2008147445A2 (en) * 2006-11-09 2008-12-04 University Of Maryland, Baltimore Use of 5,6-dimethylxanthenone-4-acetic acid as an antimicrobial agent
TWI434684B (en) * 2007-03-13 2014-04-21 Spectrum Pharmaceuticals Inc Intravesical apaziquone administration following transurethral resection
GB0800311D0 (en) * 2008-01-09 2008-02-20 Cytosystems Ltd Apparatus and method for filtering biological material
WO2011089604A2 (en) 2010-01-20 2011-07-28 Theracoat Ltd Material and method for treating internal cavities
CN102906157B (en) 2010-03-01 2016-08-24 不列颠哥伦比亚大学 Derivative hyperbranched poly glycerine
TR201807363T4 (en) * 2010-09-22 2018-06-21 Univ Texas Cancer treatment methods including targeting nqo1.
EP2734187B1 (en) 2011-07-20 2018-09-05 UroGen Pharma Ltd. Materials and method for treating internal body cavities
HUE032240T2 (en) 2011-10-24 2017-09-28 Asana Biosciences Llc Cyclohexylamines
WO2014062856A1 (en) 2012-10-16 2014-04-24 Halozyme, Inc. Hypoxia and hyaluronan and markers thereof for diagnosis and monitoring of diseases and conditions and related methods
AU2014365823B2 (en) 2013-12-16 2019-05-02 Abs Development 1, Inc. P2X3 and/or P2X2/3 compounds and methods
CN104743887B (en) * 2014-09-22 2016-03-23 巨石集团有限公司 A kind of glass fiber compound and glass fibre thereof and matrix material
KR20190053963A (en) 2016-09-30 2019-05-20 아사나 바이오사이언시스 엘엘씨 P2X3 and / or P2X2 / 3 compounds and methods
EP3924521A4 (en) 2019-02-15 2023-03-29 IncellDx, Inc. Assaying bladder-associated samples, identifying and treating bladder-associated neoplasia, and kits for use therein

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987006227A2 (en) * 1986-04-17 1987-10-22 Jouhar Akbar Jan Indoloquinone compounds
EP0393575A1 (en) * 1989-04-17 1990-10-24 G.D. Searle & Co. Neoplasia treatment compositions containing antineoplastic agent and side-effect reducing protective agent

Family Cites Families (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3993754A (en) * 1974-10-09 1976-11-23 The United States Of America As Represented By The United States Energy Research And Development Administration Liposome-encapsulated actinomycin for cancer chemotherapy
JPS54151134A (en) 1978-05-19 1979-11-28 Kureha Chem Ind Co Ltd Antitumorigenic agent
NL8001280A (en) * 1980-03-04 1981-10-01 Philips Nv LOW PRESSURE DISCHARGE LAMP.
US4615697A (en) 1983-11-14 1986-10-07 Bio-Mimetics, Inc. Bioadhesive compositions and methods of treatment therewith
US4898729A (en) 1983-12-09 1990-02-06 Euroceltique, S.A. Treatment of hypertension, compounds and compositions for antihypertension and diuresis
US4671954A (en) * 1983-12-13 1987-06-09 University Of Florida Microspheres for incorporation of therapeutic substances and methods of preparation thereof
JPS60163621A (en) 1984-02-02 1985-08-26 村上 俊文 Production of toilet seat cover made of paper
JPS60163821A (en) 1984-02-03 1985-08-26 Tdk Corp Carrier for pharmaceutical containing ferrite particle
JPS61189215A (en) 1985-02-18 1986-08-22 Teijin Ltd Oily pharmaceutical composition of 5-fluoro-2'-deoxyuridine ester
US4871542A (en) * 1987-04-30 1989-10-03 Ferring Service Center, N.V. Method and apparatus useful for delivering medicinal compositions into the bladder and urinary tract
EP0331635A3 (en) 1988-02-29 1990-02-28 The Board of Regents of the University of Texas System Preparations for treating bladder cancer
EP0338679A3 (en) 1988-03-24 1991-03-06 Genentech, Inc. Tumour necrosis factor in the treatment of bladder cancer
FR2629584B1 (en) 1988-03-31 1993-06-04 France Etat Armement STABILIZATION DEVICE FOR A LONGITUDINAL INERTIA LOW MOMENT PROJECTILE FROM A STRIPED TUBE
US5843156A (en) * 1988-08-24 1998-12-01 Endoluminal Therapeutics, Inc. Local polymeric gel cellular therapy
US5744166A (en) * 1989-02-25 1998-04-28 Danbiosyst Uk Limited Drug delivery compositions
US5216011A (en) * 1989-09-01 1993-06-01 Bristol-Myers Squibb Co. Stable solutions of mitomycin c
US5237466A (en) 1989-11-02 1993-08-17 International Business Machines Corporation Method and apparatus for programmably controlling spindle synchronization and phase among disk drives in a storage subsystem
FI84874C (en) 1989-11-02 1992-02-10 Valio Meijerien FOERFARANDE FOER INKAPSLING AV BIOLOGISKT AKTIVA AEMNEN, ENLIGT METODEN FRAMSTAELLD KAPSEL, DESS ANVAENDNING OCH FODER INNEHAOLLANDE DEN.
US5292516A (en) * 1990-05-01 1994-03-08 Mediventures, Inc. Body cavity drug delivery with thermoreversible gels containing polyoxyalkylene copolymers
US5346703A (en) * 1990-08-07 1994-09-13 Mediventures, Inc. Body cavity drug delivery with thermo-irreversible polyoxyalkylene and ionic polysaccharide gels
IT1250421B (en) 1991-05-30 1995-04-07 Recordati Chem Pharm CONTROLLED RELEASE PHARMACEUTICAL COMPOSITION WITH BIO-ADHESIVE PROPERTIES.
US5550110A (en) 1992-04-22 1996-08-27 Warner-Lambert Company Endothelin Antagonists II
JP3598049B2 (en) 1992-09-18 2004-12-08 山之内製薬株式会社 Hydrogel sustained release formulation
US5744155A (en) * 1993-08-13 1998-04-28 Friedman; Doron Bioadhesive emulsion preparations for enhanced drug delivery
ES2145102T3 (en) 1993-09-09 2000-07-01 Takeda Chemical Industries Ltd FORMULATION COMPRISING AN ANTIBACTERIAL SUBSTANCE AND AN ANTI-ULCER SUBSTANCE.
CA2135151A1 (en) 1993-11-08 1995-05-09 Mitsuhiro Wakimasu Cyclic hexapeptides, their production and use
US5405622A (en) * 1993-12-22 1995-04-11 Vernice; Joseph Gamma radiation resistant lubricating gel
TW438601B (en) * 1994-05-18 2001-06-07 Janssen Pharmaceutica Nv New mucoadhesive emulsion compositions and a process for the preparation thereof
US5811416A (en) 1994-06-06 1998-09-22 Board Of Regents The University Of Texas System Endothelin antagonist and/or endothelin synthase inhibitor in combination with a progestin, an estrogen, a cyclooxygenase inhibitor, or a nitric acid donor or substrate
US5612359A (en) 1994-08-26 1997-03-18 Bristol-Myers Squibb Company Substituted biphenyl isoxazole sulfonamides
MX9703988A (en) 1994-12-12 1998-02-28 Omeros Med Sys Inc Irrigation solution and method for inhibition of pain, inflammation and spasm.
US5749845A (en) * 1995-01-25 1998-05-12 Iotek, Inc. Delivering an agent to an organ
WO1997023456A1 (en) 1995-12-21 1997-07-03 British Technology Group Ltd. Indoloquinone derivatives as bioreductive agents
US5985312A (en) 1996-01-26 1999-11-16 Brown University Research Foundation Methods and compositions for enhancing the bioadhesive properties of polymers
CA2248076A1 (en) * 1996-03-11 1997-09-18 Richard D. Leavitt Polymeric delivery of radionuclides and radiopharmaceuticals
US5955096A (en) * 1996-06-25 1999-09-21 Brown University Research Foundation Methods and compositions for enhancing the bioadhesive properties of polymers using organic excipients
EP0815870A3 (en) 1996-06-27 2000-05-03 Takeda Chemical Industries, Ltd. Composition for prohylaxis or treatment of cerebral infarction
JP2967734B2 (en) * 1996-10-18 1999-10-25 日本電気株式会社 Thin film formation method
CA2285591A1 (en) * 1997-04-03 1998-10-08 Point Biomedical Corporation Intravesical drug delivery system
US5854382A (en) * 1997-08-18 1998-12-29 Meadox Medicals, Inc. Bioresorbable compositions for implantable prostheses
AU9082798A (en) * 1997-09-08 1999-03-29 Theramark Limited Treatment of inflammatory conditions
US6156744A (en) 1998-03-19 2000-12-05 Cancer Research Campaign Tech (London) DT-diaphorase directed anti-tumor agents
EP0950418A2 (en) 1998-04-14 1999-10-20 Takeda Chemical Industries, Ltd. Composition for preventing or treating ischemic disease
US6171610B1 (en) * 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
CA2330667A1 (en) 1998-05-05 1999-12-23 Astrazeneca Ab Mycobacterial inhibitors
IL124957A0 (en) 1998-06-16 1999-01-26 Univ Ben Gurion Active ingredient delivery systems and devices based on porous matrices
US6087396A (en) * 1998-10-05 2000-07-11 Situs Corporation Oxybutynin formulations and method of use
GB9827034D0 (en) 1998-12-10 1999-02-03 Univ Manchester Delivery formulation
JP2000256182A (en) 1999-03-03 2000-09-19 Kazuhiro Morimoto Antimicrobial formulation
ATE275727T1 (en) 1999-04-29 2004-09-15 Coley Pharm Gmbh SCREENING FOR MODULATORS OF IMMUNO-STIMULATIVE DNA FUNCTION
US7566452B1 (en) 1999-05-04 2009-07-28 New York University Cancer treatment with endothelin receptor antagonists
US6545048B1 (en) 1999-06-29 2003-04-08 California Institute Of Technology Compositions and methods of treating cancer using compositions comprising an inhibitor or endothelin receptor activity
JP2003535061A (en) 2000-05-31 2003-11-25 ワーナー−ランバート・カンパニー、リミテッド、ライアビリティ、カンパニー Conjugates of endothelin receptor antagonists and antiepileptic compounds or analgesics with pain-relieving properties
WO2002044183A2 (en) 2000-12-01 2002-06-06 Guilford Pharmaceuticals Inc. Benzoazepine and benzodiazepine derivatives and their use as parp inhibitors
US6573285B2 (en) 2000-12-21 2003-06-03 Bristol-Myers Squibb Co. Method for preventing or treating pain by administering an endothelin antagonist
US20030104976A1 (en) 2001-07-23 2003-06-05 Gudarz Davar Analgesic methods using endothelin receptor ligands
CA2466148C (en) 2001-11-01 2013-01-08 Spectrum Pharmaceuticals, Inc. Medical compositions for intravesical treatment of bladder cancer
US7973064B2 (en) 2001-11-27 2011-07-05 The Board Of Trustees Of The University Of Illinois Method and composition for potentiating an opiate analgesic
US20040009918A1 (en) 2002-05-03 2004-01-15 Hanne Nedergaard Stabilised solid compositions of modified factor VII
ATE536886T1 (en) 2002-05-03 2011-12-15 Novo Nordisk As STABILIZED SOLID COMPOSITIONS OF MODIFIED FACTOR VII
JP2006508086A (en) 2002-10-24 2006-03-09 ザ ボード オブ トラスティーズ オブ ザ ユニバーシティ オブ イリノイ Methods and compositions for preventing and treating solid tumors
PT2298815E (en) 2005-07-25 2015-07-16 Emergent Product Dev Seattle B-cell reduction using cd37-specific and cd20-specific binding molecules
RU2396953C2 (en) 2006-02-09 2010-08-20 Спектрум Фармасьютикалз, Инк. E09 and propylene glycol therapy of urinary bladder cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987006227A2 (en) * 1986-04-17 1987-10-22 Jouhar Akbar Jan Indoloquinone compounds
EP0393575A1 (en) * 1989-04-17 1990-10-24 G.D. Searle & Co. Neoplasia treatment compositions containing antineoplastic agent and side-effect reducing protective agent

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CHOUDRY G A ET AL: "A novel strategy for NQO1 (NAD(P)H:quinone oxidoreductase, EC 1.6.99.2) mediated therapy of bladder cancer based on the pharmacological properties of EO9" BRITISH JOURNAL OF CANCER 19 OCT 2001 UNITED KINGDOM, vol. 85, no. 8, 19 October 2001 (2001-10-19), pages 1137-1146, XP008014927 ISSN: 0007-0920 *
CUMMINGS JEFFREY ET AL: "Pharmacological and biochemical determinants of the antitumour activity of the indoloquinone EO9." BIOCHEMICAL PHARMACOLOGY, vol. 55, no. 3, 1 February 1998 (1998-02-01), pages 253-260, XP002237038 ISSN: 0006-2952 cited in the application *
HOLLAND AND FREI: "Cancer Medicine e.5" 2000 , B. C. DECKER INC. , HAMILTON, CANADA XP002235200 page 1543 -page 1558 *
LI DONG ET AL: "Distribution of DT-diaphorase and cytochrome P450 reductase in human bladder tissues and tumors." PROCEEDINGS OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH ANNUAL, vol. 42, March 2001 (2001-03), page 648 XP002235376 92nd Annual Meeting of the American Association for Cancer Research;New Orleans, LA, USA; March 24-28, 2001, March, 2001 ISSN: 0197-016X *
NICOLAUS B J R: "SYMBIOTIC APPROACH TO DRUG DESIGN" DECISION MAKING IN DRUG RESEARCH, XX, XX, 1983, pages 173-186, XP001111439 *
ROBERTSON N ET AL: "Factors affecting sensitivity to EO9 in rodent and human tumour cells in vitro: DT-diaphorase activity and hypoxia." EUROPEAN JOURNAL OF CANCER, vol. 30A, no. 7, 1994, pages 1013-1019, XP008015028 ISSN: 0959-8049 *
SCHELLENS JAN H M ET AL: "Phase I and pharmacologic study of the novel indoloquinone bioreductive alkylating cytotoxic drug E09." JOURNAL OF THE NATIONAL CANCER INSTITUTE (BETHESDA), vol. 86, no. 12, 1994, pages 906-912, XP008015031 ISSN: 0027-8874 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8563592B2 (en) 2001-11-01 2013-10-22 Spectrum Pharmaceuticals, Inc. Bladder cancer treatment and methods
US8648108B2 (en) 2001-11-01 2014-02-11 Spectrum Pharmaceuticals, Inc. Medical compositions for intravesical treatment of bladder cancer
US9295666B2 (en) 2001-11-01 2016-03-29 Spectrum Pharmaceuticals, Inc. Bladder cancer treatment and methods
WO2006105507A2 (en) * 2005-03-31 2006-10-05 Spectrum Pharmaceuticals, Inc. Indoloquinone tumor radiation sensitization
WO2006105507A3 (en) * 2005-03-31 2007-03-01 Spectrum Pharmaceuticals Inc Indoloquinone tumor radiation sensitization
WO2007092964A1 (en) * 2006-02-09 2007-08-16 Spectrum Pharmaceuticals, Inc. Bladder cancer treatment by using e09 and propylene glycol
WO2021008520A1 (en) * 2019-07-15 2021-01-21 深圳艾欣达伟医药科技有限公司 Stable ast-3424 injection preparation and preparation method

Also Published As

Publication number Publication date
EP1864660A2 (en) 2007-12-12
US20030133954A1 (en) 2003-07-17
EP1864660B1 (en) 2010-12-29
US7977369B2 (en) 2011-07-12
CA2466148C (en) 2013-01-08
US8648108B2 (en) 2014-02-11
EP1864660A3 (en) 2008-04-23
JP4317452B2 (en) 2009-08-19
CA2466148A1 (en) 2003-05-08
PT1864660E (en) 2011-03-31
ES2341922T3 (en) 2010-06-29
US20070010570A1 (en) 2007-01-11
US20120252861A1 (en) 2012-10-04
DK2060259T3 (en) 2010-05-17
WO2003037314A3 (en) 2003-10-16
CA2789114A1 (en) 2003-05-08
DE60238798D1 (en) 2011-02-10
DE60235214D1 (en) 2010-03-11
AU2002350115A1 (en) 2003-05-12
US6894071B2 (en) 2005-05-17
EP1439835A2 (en) 2004-07-28
PT2060259E (en) 2010-04-26
CA2789114C (en) 2014-03-25
EP2060259A1 (en) 2009-05-20
EP2060259B1 (en) 2010-01-20
US20090076123A1 (en) 2009-03-19
CY1111252T1 (en) 2015-08-05
HK1132181A1 (en) 2010-02-19
DK1864660T3 (en) 2011-02-21
ATE493126T1 (en) 2011-01-15
US20050215615A1 (en) 2005-09-29
JP2005532986A (en) 2005-11-04
US20080166402A1 (en) 2008-07-10
ATE455545T1 (en) 2010-02-15
ES2384208T3 (en) 2012-07-02

Similar Documents

Publication Publication Date Title
US8648108B2 (en) Medical compositions for intravesical treatment of bladder cancer
Choudry et al. A novel strategy for NQO1 (NAD (P) H: quinone oxidoreductase, EC 1.6. 99.2) mediated therapy of bladder cancer based on the pharmacological properties of EO9
EP0738152B1 (en) Use of 2-hydroxy-5-phenylazobenzoic acid derivatives as colon cancer chemopreventative and chemotherapeutic agents
EP1047416B1 (en) Pharmaceutical compositions comprising disulfiram for inhibiting angiogenesis
Guichard et al. CPT-11 converting carboxylesterase and topoisomerase I activities in tumour and normal colon and liver tissues
Nicotera et al. Cytosolic‐free Ca2+ and cell killing in hepatoma 1c1c7 cells exposed to chemical anoxia
US9295666B2 (en) Bladder cancer treatment and methods
Boyer et al. Lysosomes, lysosomal enzymes, and cancer
JP2005537277A (en) Antitumor compounds and their therapeutic use
Jaffar et al. Targeting hypoxia with a new generation of indolequinones
US5905073A (en) Use of 2-hydroxy-5-phenylazobenzoic acid derivatives as colon cancer chemopreventative and chemotherapeutic agents
US20110288472A1 (en) Bladder cancer treatment and methods
Block et al. Evaluation of aminoglutethimide in stage D prostate cancer: an assessment of efficacy and toxicity in patients with tumors refractory to hormonal therapy.
Ning et al. Preparation and characterization of EP-liposomes and Span 40-niosomes
Roy et al. Prophylactic and therapeutic role of catechin-loaded poly (D, L-lactic-co-glycolic acid) nanocapsules in gastric ulcer by in vitro and in vivo approach
Tan et al. Relation of malignant tumor to prostacyclin and thromboxane

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC 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 MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2466148

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003539658

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2002786643

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002786643

Country of ref document: EP