US20030004093A1 - Method for identifying compounds for inhibition of neoplastic lesions - Google Patents

Method for identifying compounds for inhibition of neoplastic lesions Download PDF

Info

Publication number
US20030004093A1
US20030004093A1 US10/040,776 US4077602A US2003004093A1 US 20030004093 A1 US20030004093 A1 US 20030004093A1 US 4077602 A US4077602 A US 4077602A US 2003004093 A1 US2003004093 A1 US 2003004093A1
Authority
US
United States
Prior art keywords
compounds
cells
compound
inhibition
apoptosis
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/040,776
Inventor
Gary Piazza
Rifat Pamukcu
W. Thompson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
OSI Pharmaceuticals LLC
Original Assignee
Piazza Gary A.
Rifat Pamukcu
Thompson W. Joseph
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/866,027 external-priority patent/US5858694A/en
Application filed by Piazza Gary A., Rifat Pamukcu, Thompson W. Joseph filed Critical Piazza Gary A.
Priority to US10/040,776 priority Critical patent/US20030004093A1/en
Publication of US20030004093A1 publication Critical patent/US20030004093A1/en
Assigned to OSI PHARMACEUTICALS, INC. reassignment OSI PHARMACEUTICALS, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: CELL PATHWAYS, INC.
Assigned to OSI PHARMACEUTICALS, INC. reassignment OSI PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CELL PATHWAYS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/44Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving esterase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/533Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving isomerase

Definitions

  • This invention provides a method for identifying compounds potentially useful for the treatment and prevention of pre-cancerous and cancerous lesions in mammals.
  • This application is a continuation-in-part of U.S. patent application Ser. No. 08/866,027 to Piazza et al. Filed May 30, 1997.
  • Familial adenomatous polyposis (“FAP”) is an inherited disease where the victim's colon contains many polyps or adenomas virtually uncountable in most instances. Because such patients develop so many polyps or adenomas each of which has a significant risk of developing into a cancer—the typical treatment is surgical removal of the colon.
  • NSAID nonsteroidal anti-inflammatory drug
  • sulindac would cause colonic polyps (a type of pre-cancerous lesion) to regress and prevent their recurrence when that drug was administered to patients with FAP. Waddell's experience with sulindac in FAP patients was confirmed in several subsequent studies.
  • Prostaglandin (“PG”) synthesis inhibition results from the inhibition of cyclooxygenase (COX) caused by NSAIDs.
  • COX cyclooxygenase
  • a common benefit of NSAIDs is the reduction of inflammation, which is known to be caused by the reduction of PG levels. Since NSAIDs are known to inhibit COX, which inhibits PG synthesis, it is widely believed that the regression of colonic polyps is attributed to this property.
  • NSAID therapy In significant numbers of cases, NSAID therapy must be stopped due to the severity of those side effects and other potentially lethal side effects. Furthermore, the severity of such side effects increases with age, probably because natural PG levels in gastric mucosa falls with age.
  • useful compounds for treating neoplastic lesions should desirably inhibit neoplastic cell growth, but should not inhibit COX.
  • rational drug discovery methods are being applied in the pharmaceutical industry to improve methods for identifying clinically useful compounds.
  • rational drug discovery methods relate to a “lock and key” concept whereby structural relationships between a therapeutic target molecule (lock) and pharmaceutical compounds (key) are defined.
  • Such methods are greatly enhanced by specialty computer software that accesses databases of compounds to identify likely geometric fits with the target molecule.
  • the target may be an enzyme, a protein, a membrane or nuclear receptor, or a nucleic acid sequence, for example.
  • neoplasia In complex diseases, such as neoplasia, scientists have identified a number of potential targets. However, many of the drugs available for the treatment of neoplasia are non-specific and toxic to normal tissues, and are not indicated for precancer and used only when neoplastic cells progress to cancer. Greater understanding of the mechanisms involved in cancer may lead scientists on the path towards designing more specific antineoplastic drugs—drugs that can safely be administered earlier in the disease process.
  • This invention relates to a novel in vitro method for screening test compounds for their ability to treat and prevent neoplasia, especially pre-cancerous lesions, safely.
  • the present invention provides a method for identifying test compounds that can be used to treat and prevent neoplasia, including precancerous lesions, with minimal side effects associated with COX inhibition and other non-specific interactions.
  • the screening method involves determining the COX inhibition activity of a test compound. Because the inventors have discovered a relationship between inhibition of cancer and inhibition of phosphodiesterase Type-5 isoenzyme (“PDE5”), this invention includes determining the PDE5 inhibition activity of the compound. Preferably, the screening method of this invention farther includes determining whether the compounds inhibit the growth of tumor cells in a cell culture.
  • PDE5 phosphodiesterase Type-5 isoenzyme
  • the screening method of this invention involves determining the COX inhibition activity of the compound, determining the PDE5 inhibition activity of the compound and determining whether the compound induces apoptosis in tumor cells.
  • FIG. 1 illustrates the effect of the sulfide derivative of sulindac and the sulfone derivative of sulindac (a.k.a. exisulind) on purified cyclooxygenase activity.
  • FIG. 2 illustrates the effects of test compounds B and E on COX inhibition.
  • FIG. 3 illustrates the inhibitory effects of sulindac sulfide and exisulind on PDE-4 and PDE5 purified from cultured tumor cells.
  • FIG. 4 illustrates the effects of sulindac sulfide on cyclic nucleotide levels in HT-29 cells.
  • FIG. 5 illustrates the phosphodiesterase inhibitory activity of compound B.
  • FIG. 6 illustrates the phosphodiesterase inhibitory activity of compound E.
  • FIG. 7 illustrates the effects of sulindac sulfide and exisulind on apoptosis and necrosis of HT-29 cells.
  • FIG. 8 illustrates the effects of sulindac sulfide and exisulind on HT-29 cell growth inhibition and apoptosis induction as determined by DNA fragmentation.
  • FIG. 9 illustrates the apoptosis inducing properties of compound E.
  • FIG. 10 illustrates the apoptosis inducing properties of compound B.
  • FIG. 11 illustrates the effects of sulindac sulfide and exisulind on tumor cell growth.
  • FIG. 12 illustrates the growth inhibitory and apoptosis-inducing activity of sulindac sulfide and control (DMSO).
  • FIG. 13 illustrates the growth inhibitory activity of compound E.
  • FIG. 14 illustrates the inhibition of pre-malignant, neoplastic lesions in mouse mammary gland organ culture by sulindac metabolites.
  • the method of this invention is useful to identify compounds that can be used to treat or prevent neoplasms, and which are not characterized by the serious side effects of conventional NSAIDs.
  • Cancer and precancer may be thought of as diseases that involve unregulated cell growth.
  • Cell growth involves a number of different factors. One factor is how rapidly cells proliferate, and another involves how rapidly cells die. Cells can die either by necrosis or apoptosis depending on the type of environmental stimuli. Cell differentiation is yet another factor that influences tumor growth kinetics. Resolving which of the many aspects of cell growth is affected by a test compound is important to the discovery of a relevant target for pharmaceutical therapy. Screening assays based on this selectivity can be combined with tests to determine which compounds having growth inhibiting activity.
  • This invention is the product of several important discoveries.
  • PDE phosphodiesterase
  • desirable compounds for treating neoplastic lesions selectively inhibit Type 5 isoenzyme form of phosphodiesterase (“PDE5”) (EC 3.1.4.17).
  • PDE5 is one of at least seven isoenzymes of phosphodiesterase.
  • PDE5 is unique in that it selectively degrades cyclic GMP, while the other types of PDE are either non-selective or degrade cyclic AMP.
  • desirable compounds do not substantially inhibit other phosphodiesterase types.
  • a preferred embodiment of the present invention involves determining the cyclooxygenase inhibition activity of a given compound, and determining the PDE5 inhibition activity of the compound.
  • the test compounds are scored for their probable ability to treat neoplastic lesions either directly by assessing their activities against specific cutoff values or indirectly by comparing their activities against known compounds useful for treating neoplastic lesions.
  • a standard compound that is known to be effective for treating neoplastic lesions without causing gastric ulceration is 5-fluoro-2-methyl-1-(p-methylsulfonylbenzylidene)-3-indenylacetic acid (“exisulind”).
  • useful compounds for comparative purposes include those that are known to inhibit COX, such as indomethacin and the sulfide metabolite of sulindac: 5-fluoro-2-methyl-1-(p-methylsulfinylbenzylidene)-3-indenylacetic acid (“sulindac sulfide”).
  • Other useful compounds for comparative purposes include those that are known to inhibit PDE5, such as 1-(3-chloroanilino)-4-phenyphthalazine (“MY5445”).
  • test compound is clearly determined to be a promising candidate if it performs better than or comparable to exisulind and does not inhibit COX.
  • desirable compounds are those that inhibit PDE5 and inhibit cell growth and induce apoptosis, but do not inhibit COX at pharmacologically accepted doses.
  • precancerous lesion includes syndromes represented by abnormal neoplastic, including dysplastic, changes of tissue. Examples include dysplastic growths in colonic, breast, prostate or lung tissues, or conditions such as dysplastic nevus syndrome, a precursor to malignant melanoma of the skin.
  • Examples also include, in addition to dysplastic nevus syndromes, polyposis syndromes, colonic polyps, precancerous lesions of the cervix (i.e., cervical dysplasia), esophagus, lung, prostatic dysplasia, prostatic intraneoplasia, breast and/or skin and related conditions (e.g., actinic keraosis), whether the lesions are clinically identifiable or not.
  • cancer refers to lesions which are cancerous. Examples include malignant melanomas, breast cancer, prostate cancer and colon cancer. As used herein, the terms “neoplasia” and “neoplasms” refer to both cancerous and pre-cancerous lesions.
  • PG represents prostaglandin
  • PS represents prostaglandin synthetase
  • PGE 2 represents prostaglandin E 2
  • PDE represents phosphodiesterase
  • COX represents cyclooxygenase
  • RIA represents—radioimmunoassay.
  • PDE5 refers to that enzyme and any of its isoforms that exhibit cGMP specific hydrolytic enzyme activities and high affinity cGMP binding.
  • COX inhibition can be determined by either of two methods.
  • One method involves measuring PGE 2 secretion by intact HL-60 cells following exposure to the compound being screened.
  • the other method involves measuring the activity of purified cyclooxygenases (COXs) in the presence of the compound. Both methods involve protocols previously described in the literature.
  • PGE 2 prostaglandin E 2
  • EIA enzyme immunoassay
  • Suitable cells include those which make an abundance of PG, such as HL-60 cells.
  • HL-60 cells are human promyelocytes that are differentiated with DMSO in mature granulocytes.
  • HL-60 Human Promyelocytic Leukemia Cells (HL-60) After Induction of Differentiation By Dimethylsulfoxide”, J. Exp. Med., 149:969-974, 1979). These differentiated cells produce PGE 2 after stimulation with a calcium ionophore A23187 (see, Kargman, S., Prasit, P. and Evans, J. F., “Translocation of HL-60 Cell 5-Lipoxygenase”, J. Biol. Chem., 266: 23745-23752, 1991). HL-60 are available from the American Type Culture Collection (ATCC:CCL240).
  • the differentiated HL-60 cells (3 ⁇ 10 6 cells/ml) can be incubated for 15 min at 37° C. in the presence of the compounds tested at the desired concentration. Cells are then stimulated by A23187 (5 ⁇ 10 ⁇ 6 M) for 15 min. PGE 2 secreted into the external medium is measured as described above.
  • COX-I and COX-2 Two different forms of cyclooxygenase (COX-I and COX-2) have been reported in the literature to regulate prostaglandin synthesis. It is known that COX-2 represents the inducible form of COX while COX-I represents a constitutive form. COX-I activity can be measured using the method described by Mitchell et al. (“Selectivity of Nonsteroidal Anti-inflammatory Drugs as Inhibitors of Constitutive and Inducible Cyclooxygenase,” Proc. Natl. Acad. Sci.
  • COX-2 activity can be measured using COX-2 purified from sheep placenta as described by Mitchell et al., 1993, supra.
  • the cyclooxygenase inhibitory activity of a drug can be determined by methods known in the art. For example, Boopathy & Balasubramanian, 1988, supra, described a procedure in which prostaglandin H synthase 1 (Cayman Chemical, Ann Arbor, Mich.) is incubated at 37° C. for 20 min with 100 ⁇ M arachidonic acid (Sigma Chemical Co.), cofactors (such as 1.0 mM glutathione, 1.0 mM hydroquinone, 0.625 ⁇ M hemoglobin and 1.25 mM CaCl 2 in 100 mM Tris-HCl, pH 7.4) and the drug to be tested. Following incubation, the reaction can be terminated with trichloroacetic acid. Enzymatic activity can then be measured spectrophotometrically at 530 nm after stopping the reaction by adding thiobarbituric acid and malonaldehyde.
  • the amount of inhibition is determined by comparing the activity of the cyclooxygenase in the presence and absence of the test compound. Residual or no COX inhibitory activity (i.e., less than about 25%) at a concentration of about 100 ⁇ M is indicative that the compound should be evaluated further for usefulness for treating neoplasia. Preferably, the IC 50 concentration should be greater than 1000 ⁇ M for the compound to be further considered potential use.
  • Compounds can be screened for inhibitory effect on phosphodiesterase activity using either the enzyme isolated from any tumor cell line such as HT-29 or SW-480, or recombinant HS-PDE5, for example, or measuring cyclic nucleotide levels in whole cells.
  • Phosphodiesterase activity can be determined using methods known in the art, such as a method using radioactive 3 H cyclic GMP (cGMP)(cyclic 3′,5′-guanosine monophosphate) as the substrate for PDE5 enzyme.
  • cGMP radioactive 3 H cyclic GMP
  • a solution of defined substrate 3 H-cGMP specific activity (0.2 ⁇ M; 100,000 cpm; containing 40 mM Tris-HCl (pH 8.0), 5 mM MgCl 2 and 1 mg/ml BSA) is mixed with the drug to be tested in a total volume of 400 ⁇ l.
  • the mixture is incubated at 30° C. for 10 minutes with partially purified PDE5 isolated from HT-29 cells. Reactions are terminated, for example, by boiling the reaction mixture for 75 seconds. After cooling on ice, 100 ⁇ l of 0.5 mg/ml snake venom (O. Hannah venom available from Sigma) is added and incubated for 10 min at 30° C.
  • This reaction is then terminated by the addition of an alcohol, e.g. 1 ml of 100% methanol.
  • Assay samples are applied to a anion chromatography column (1 ml Dowex, from Aldrich) and washed with 1 ml of 100% methanol.
  • the amount of radioactivity in the breakthrough and the wash from the columns in then measured with a scintillation counter.
  • the degree of PDE5 inhibition is determined by calculating the amount of radioactivity in drug-treated reactions and comparing against a control sample (a reaction mixture lacking the tested compound).
  • the ability for desirable compounds to inhibit PDE5 is reflected by an increase in cGMP in neoplastic cells exposed to a compound being screened.
  • the amount of PDE5 activity can be determined by assaying for the amount of cyclic GMP in the extract of treated cells using radioimmunoassay (RIA).
  • RIA radioimmunoassay
  • HT-29 or SW-480 cells are plated and grown to confluency.
  • the test compound is then incubated with the cell culture at a concentration of compound between about 200 ⁇ M to about 200 pM. About 24 to 48 hours thereafter, the culture media is removed from the cells, and the cells are solubilized. The reaction is stopped by using 0.2N HCl/50%-MeOH. A sample is removed for protein assay.
  • Cyclic GMP is purified from the acid/alcohol extracts of cells using anion-exchange chromatography, such as a Dowex column.
  • the cGMP is dried, acetylated according to published procedures, such as using acetic anhydride in triethylamine, (Steiner, A. L., Parker, C. W., Kipnis, D. M., J. Biol Chem., 247(4): 1106-13, 1971, which is incorporated herein by reference).
  • the acetylated cGMP is quantitated using radioimmunoassay procedures (Harper, J., Brooker, G., Advances in Nucleotide Research, 10:1-33, 1979, which is incorporated herein by reference).
  • Iodinated ligands (tyrosine metheyl ester) of derivatized cyclic GMP are incubated with standards or unknowns in the presence of antisera and appropriate buffers.
  • Antiserum may be produced using cyclic nucleotide-haptene directed techniques. The antiserum is from sheep injected with succinyl-cGMP-albumin conjugates and diluted 1/20,000. Dose-interpolation and error analysis from standard curves are applied as described previously (Seibert, A. F., Thompson, W. J., Taylor, A., Wilbourn, W. H., Barnard, J. and Haynes, J., J. Applied Physio., 79:389-395, 1992, which is incorporated hereinby reference).
  • the culture media may be acidified, frozen ( ⁇ 70° C.) and also analyzed for cGMP and cAMP.
  • the change in the ratio of the two cyclic nucleotides may be a more accurate tool for evaluating desirable PDE5 inhibition activity of test compounds, rather than measuring only the absolute value of cGMP, only PDE5 inhibition, or only the absolute value of cGMP.
  • the ratio of cGMP content/cAMNP content is in the 0.03-0.05 range (i.e., 300-500 fmol/mg protein cGMP content over 6000-8000 fmol/mg protein cAMP content).
  • that ratio increases several fold (preferably at least about a three-fold increase) as the result of an initial increase in cyclic GMP and the later decrease in cyclic AMP.
  • particularly desirable compounds achieve an initial increase in cGMP content in treated neoplastic cells to a level of cGMP greater than about 500 fmol/mg protein.
  • particularly desirable compounds cause the later decrease in cAMP content in treated neoplastic cells to a level of cAMP less than about 4000 fmol/mg protein.
  • cyclic nucleotides are purified from acid/alcohol extracts of cells using anion-exchange chromatography, dried, acetylated according to published procedures and quantitated using radioimmunoassay procedures. Iodinated ligands of derivatized cyclic AMP and cyclic GMP are incubated with standards or unknowns in the presence of specific antisera and appropriate buffers.
  • Verification of the cyclic nucleotide content may be obtained by determining the turnover or accumulation of cyclic nucleotides in intact cells.
  • 3 H-adenine prelabeling is used according to published procedures (Whalin M. E., R. L. Garrett Jr., W. J. Thompson, and S. J. Strada, “Correlation of cell-free brain cyclic nucleotide phosphodiesterase activities to cyclic AMP decay in intact brain slices”, Sec. Mess. and Phos. Protein Research, 12:31 1-325, 1989, which is incorporated herein by reference).
  • the procedure measures flux of labeled ATP to cyclic AMP and can be used to estimate intact cell adenylate cyclase or cyclic nucleotide phosphodiesterase activities depending upon the specific protocol. Cyclic GMP accumulation was too low to be studied with intact cell prelabeling according to published procedures (Reynolds, P. E., S. J. Strada and W. J. Thompson, “Cyclic GMP accumulation in pulmonary microvascular endothelial cells measured by intact cell prelabeling,” Life Sci., 60:909-918, 1997, which is incorporated herein by reference).
  • the PDE5 inhibitory activity of a test compound can also be determined from a tissue sample.
  • Tissue samples such as mammalian (preferably rat) liver, are collected from subjects exposed to the test compound. Briefly, a sample of tissue is homogenized in 500 ⁇ l of 6% TCA. A known amount of the homogenate is removed for protein analysis. The remaining homogenate is allowed to sit on ice for 20 minutes to allow for the protein to precipitate. Next, the homogenate is centrifuged for 30 minutes at 15,000 g at 4° C. The supernatant is recovered and the pellet recovered. The supernatant is washed four times with five volumes of water saturated diethyl ether. The upper ether layer is discarded between each wash.
  • the aqueous ether extract is dried in a speed vac. Once dried, the sample can be frozen for future use, or used immediately.
  • the dried extract is dissolved in 500 ⁇ l of assay buffer.
  • the amount of PDE5 inhibition is determined by assaying for the amount of cyclic nucleotides using an enzyme immunoassay (EIA), such as the Biotrak EIA system acetylation protocol (available from Amersham, Arlington Heights, Ill., USA). Alternatively, RIA procedures as detailed above may be used.
  • EIA enzyme immunoassay
  • the amount of inhibition is determined by comparing the activity of PDE5 in the presence and absence of the test compound. Inhibition of PDE5 activity is indicative that the compound is useful for treating neoplasia. Significant inhibitory activity greater than that of the benchmark, exisulind, preferably greater than 50% at a concentration of 10 ⁇ M or below, is indicative that a compound should be further evaluated for antineoplastic properties. Preferably, the IC 50 value for PDE5 inhibition should be less than 50 ⁇ M for the compound to be further considered for potential use.
  • the screening method of the present invention involves further determining whether the compound reduces the growth of tumor cells.
  • Various cell lines can be used in the sample depending on the tissue to be tested.
  • these cell lines include: SW-480—colonic adenocarcinoma; HT-29—colonic adenocarcinoma, A-427—lung adenocarcinoma carcinoma; MCF-7—breast adenocarcinoma; and UACC-375—melanoma line; and DU145—prostrate carcinoma. Cytotoxicity data obtained using these cell lines are indicative of an inhibitory effect on neoplastic lesions. These cell lines are well characterized. and are used by the United States National Cancer Institute in their screening program for new anti-cancer drugs.
  • a compound's ability to inhibit tumor cell growth can be measured using the HT-29 human colon carcinoma cell line obtained from ATCC (Bethesda, Md.).
  • HT-29 cells have previously been characterized as relevant colon tumor cell culture model (Fogh, J., and Trempe, G. In: Human Tumor Cells in Vitro, J. Fogh (eds.), Plenum Press, New York, pp. 115-159, 1975).
  • HT-29 cells are maintained in RPMI media supplemented with 5% fetal serum (Gemini Bioproducts, Inc., Carlsbad, Calif.) and 2 mm glutamine, and 1% antibiotic-antimycotic in a humidified atmosphere of 95% air and 5% CO 2 at 37° C.
  • HT-29 cells are plated at a density of 500 cells/well in 96 well microtiter plates and incubated for 24 hours at 37° C. prior to the addition of compound. Each determination of cell number involved six replicates. After six days in culture, the cells are fixed by the addition of cold trichloroacetic acid to a final concentration of 10% and protein levels are measured using the sulforhodamine B (SRB) colorimetric protein stain assay as previously described by Skehan, P., Storeng, R., Scudiero, D., Monks, A., McMahon, J., Vistica, D., Warren, J. T., Bokesch, H., Kenney, S., and Boyd, M. R., “New Colorimetric Assay For Anticancer-Drug Screening,” J. Natl. Cancer Inst. 82: 1107-1112, 1990, which is incorporated herein by reference.
  • SRB sulforhodamine B
  • SRB assay In addition to the SRB assay, a number of other methods are available to measure growth inhibition and could be substituted for the SRB assay. These methods include counting viable cells following trypan blue staining, labeling cells capable of DNA synthesis with BrdU or radiolabeled thymidine, neutral red staining of viable cells, or MTT staining of viable cells.
  • IC 50 value is determined and used for comparative purposes. This value is equivalent to the concentration of drug needed to inhibit tumor cell growth by 50% relative to the control. Preferably, the IC 50 value should be less than 100 ⁇ M for the compound to be considered further for potential use for treating neoplastic lesions.
  • the screening method of the present invention further involves determining whether the compound induces apoptosis in cultures of tumor cells.
  • necrosis and apoptosis Two distinct forms of cell death may be described by morphological and biochemical criteria: necrosis and apoptosis. Necrosis is accompanied by increased permeability of the plasma membrane; the cells swell and the plasma membrane ruptures within minutes. Apoptosis is characterized by membrane blebbing, condensation of cytoplasm and the activation of endogenous endonucleases.
  • apoptosis is the most common form of eukaryotic cell death. It occurs naturally during normal tissue turnover and during embryonic development of organs and limbs. Apoptosis also is induced by cytotoxic T-lymphocytes and natural killer cells, by ionizing radiation and certain chemotherapeutic drugs. Inappropriate regulation of apoptosis is thought to play an important role in many pathological conditions including cancer, AIDS, Alzheimer disease, etc. Compounds can be screened for induction of apoptosis using cultures of tumor cells maintained under conditions as described above. Treatment of cells with test compounds involves either pre- or post-confluent cultures and treatment for two to seven days at various concentrations.
  • Apoptotic cells are measured in both the attached and “floating” compartments of the cultures. Both compartments are collected by removing the supernatant, trypsinizing the attached cells, and combining both preparations following a centrifugation wash step (10 minutes, 2000 rpm).
  • the protocol for treating tumor cell cultures with sulindac and related compounds to obtain a significant amount of apoptosis has been described in the literature. (See, Piazza, G. A., et al., Cancer Research, 55:3110-16, 1995, which is incorporated herein by reference).
  • the novel features include collecting both floating and attached cells, identification of the optimal treatment times and dose range for observing apoptosis, and identification of optimal cell culture conditions.
  • cultures can be assayed for apoptosis and necrosis by florescent microscopy following labeling with acridine orange and ethidium bromide.
  • the method for measuring apoptotic cell number has previously been described by Duke & Cohen, “Morphological And Biochemical Assays Of Apoptosis,” Current Protocols In Immunology , Coligan et al., eds., 3.17.1-3.17.16 (1992, which is incorporated herein by reference).
  • floating and attached cells can be collected by trypsinization and washed three times in PBS. Aliquots of cells can be centrifuged. The pellet can then be resuspended in media and a dye mixture containing acridine orange and ethidium bromide prepared in PBS and mixed gently. The mixture can then be placed on a microscope slide and examined.
  • Apoptosis can also be quantified by measuring an increase in DNA fragmentation in cells which have been treated with test compounds.
  • Commercial photometric EIA for the quantitative in vitro determination of cytoplasmic histone-associated-DNA-fragments (mono- and oligonucleosomes) are available (Cell Death Detection ELISA okys , Cat. No. 1,774,425, Boehringer Mannheim).
  • the Boehringer Mannheim assay is based on a sandwich-enzyme-immunoassay principle using mouse monoclonal antibodies directed against DNA and histones, respectively. This allows the specific determination of mono-and oligonucleosomes in the cytoplasmatic fraction of cell lysates.
  • apoptosis is measured in the following fashion.
  • the sample (cell-lysate) is placed into a streptavidin-coated microtiter plate (“MTP”).
  • MTP streptavidin-coated microtiter plate
  • a mixture of anti-histone-biotin and anti-DNA peroxidase conjugate are added and incubated for two hours.
  • the anti-histone antibody binds to the histone-component of the nucleosomes and simultaneously fixes the immunocomplex to the streptavidin-coated MTP via its biotinylation.
  • the anti-DNA peroxidase antibody reacts with the DNA component of the nucleosomes.
  • Peroxidase is determined photometrically with ABTS7 (2,2′-Azido-[3 -ethylbenzthiazolin-suffonate])* as substrate.
  • SW-480 colon adenocarcinoma cells are plated in a 96-well MTP at a density of 10,000 cells per well. Cells are then treated with test compound, and allowed to incubate for 48 hours at 37° C. After the incubation, the MTP is centrifuged and the supernatant is removed. The cell pellet in each well is then resuspended in lysis buffer for 30 minutes. The lysates are then centrifuged and aliquots of the supernatant (i.e. cytoplasmic fraction) are transferred into streptavidin coated MTP. Care is taken not to shake the lysed pellets (i.e. cell nucleii containing high molecular weight, unfragmented DNA) in the MTP. Samples are then analyzed.
  • EC 50 values may also be determined by evaluating a series of concentrations of the test compound.
  • apoptosis i.e., greater than 2 fold stimulation at a concentration of 100 ⁇ M
  • the EC 50 value for apoptotic activity should be less than 100 ⁇ M for the compound to be further considered for potential use for treating neoplastic lesions.
  • EC 50 is herein defined as the concentration that causes 50% induction of apoptosis relative to vehicle treatment.
  • Test compounds identified by the above methods can be tested for antineoplastic activity by their ability to inhibit the incidence of preneoplastic lesions in a mammary gland organ culture system.
  • This mouse mammary gland organ culture technique has been successfully used by other investigators to study the effects of known antineoplastic agents such as NSAIDs, retinoids, tamoxifen, selenium, and certain natural products, and is useful for validation of the screening method of the present invention.
  • female BALB/c mice can be treated with a combination of estradiol and progesterone daily, in order to prime the glands to be responsive to hormones in vitro.
  • the animals are sacrificed and thoracic mammary glands are excised aseptically and incubated for ten days in growth media supplemented with insulin, prolactin, hydrocortisone, and aldosterone.
  • DMBA 7,12-dimethylbenz(a)anthracene
  • Fully developed glands are then deprived of prolactin, hydrocortisone, and aldosterone, resulting in the regression of the glands but not the premalignant lesions.
  • test compound is dissolved in DMSO and added to the culture media for the duration of the culture period.
  • the glands were fixed in 10% formalin. stained with alum carmine, and mounted on glass slides.
  • the incidence of forming mammary lesions is the ratio of the glands with mammary lesions and glands without lesions.
  • the incidence of mammary lesions in test compound treated glands is compared with that of the untreated glands.
  • the extent of the area occupied by the mammary lesions can be quantitated by projecting an image of the gland onto a digitation pad.
  • the area covered by the gland is traced on the pad and considered as 100% of the area.
  • the space covered by each of the unregressed structures is also outlined on the digitization pad and quantitated by the computer.
  • test compounds were examined in the various protocols and screened for potential use in treating neoplasia. The results of these tests are reported below.
  • the test compounds are hereinafter designated by a letter code that corresponds to the following:
  • FIG. 1 shows the effect of various concentrations of either sulindac sulfide or exisulind on purified cyclooxygenase (Type 1) activity. Cyclooxygenase activity was determined using purified cyclooxygenase from ram seminal vesicles as described previously (Mitchell et al, supra). The IC-50 value for sulindac sulfide was calculated to be approximately 1.76 ⁇ M, while that for exisulind was greater than 10,000 ⁇ M.
  • FIG. 2 shows the effect of test compounds B and E on COX inhibition.
  • COX activity was determined as for the compounds shown in FIG. 1. The data show that both test compound B and E do not significantly inhibit COX-I.
  • TABLE 1 Cyclooxygenase inhibitory activity among a series of compounds % Inhibition at 100 ⁇ M Reference compounds Indomethacin 95 MY5445 94 Sulindac sulfide 97 Exisulind ⁇ 25 Test compounds A ⁇ 25 B ⁇ 25 C 87 D ⁇ 25 E ⁇ 25
  • FIG. 3 shows the effect of various concentrations of sulindac sulfide and exisulind on either PDE-4 or PDE5 activity purified from human colon HT-29 cultured tumor cells, as described previously (W. J. Thompson et al., supra).
  • the IC 50 value of sulindac sulfide for inhibition of PDE4 was 41 ⁇ M, and for inhibition of PDE5 was 17 ⁇ M.
  • the IC 50 value of exisulind for inhibition of PDE4 was 181 ⁇ M, and for inhibition of PDE5 was 56 ⁇ M.
  • FIG. 4 shows the effects of sulindac sulfide on either cGMP or cAMP production as determined on cultured HT-29 cells in accordance with the assay of section 2.B., supra.
  • HT-29 cells were treated with sulindac sulfide for 30 minutes and cGMP or cAMP was measured by conventional radioimmunoassay method.
  • sulindac sulfide increased the levels of cGMP by greater than 50% with an EC 50 value of 7.3 ⁇ M (top).
  • Levels of cAMP were unaffected by treatment, although a known PDE4 inhibitor, rolipram, increased cAMP (bottom).
  • the data demonstrate the pharmacological significance of inhibiting PDE5, relative to PDE4.
  • FIG. 5 shows the effect of the indicated dose of test compound B on either PDE5 or PDE4 isozymes of phosphodiesterase.
  • the calculated IC 50 value for PDE5 was 18 ⁇ M and 58 ⁇ M for PDE4.
  • FIG. 6 shows the effect of the indicated dose of test compound E on either PDE4 or PDE5.
  • the calculated IC 50 value was 0.08 ⁇ M for PDE5 and greater than 25 ⁇ M for PDE4.
  • FIG. 7 shows the effects of sulindac sulfide and exisulind on apoptotic and necrotic cell death.
  • HT-29 cells were treated for six days with the indicated dose of either sulindac sulfide or exisulind. Apoptotic and necrotic cell death was determined previously (Duke and Cohen, In: Current Protocols in Immunology, 3.17.1-3.17.16, New York, John Wiley and Sons, 1992). The data shows that both sulindac sulfide and exisulind are capable of causing apoptotic cell death without inducing necrosis. All data were collected from the same experiment.
  • FIG. 8 shows the effect of sulindac sulfide and sulfone on tumor growth inhibition and apoptosis induction as determined by DNA fragmentation.
  • Top figure growth inhibition (open symbols, right axis) and DNA fragmentation (closed symbols, left axis) by exisulind.
  • Bottom figure growth inhibition (open symbols) and DNA fragmentation (closed symbols) by sulindac sulfide. Growth inhibition was determined by the SRB assay after six days of treatment. DNA fragmentation was determined after 48 hours of treatment. All data was collected from the same experiment.
  • FIG. 9 shows the apoptosis inducing properties of compound E.
  • HT-29 colon adenocarcinoma cells were treated with the indicated concentration of compound E for 48 hours and apoptosis was determined by the DNA fragmentation assay.
  • the calculated EC 50 value was 0.05 ⁇ M.
  • FIG. 10 shows the apoptosis inducing properties of compound B.
  • HT-29 colon adenocarcinoma cells were treated with the indicated concentration of compound B for 48 hours and apoptosis was determined by the DNA fragmentation assay. The calculated EC 50 value was approximately 175 ⁇ M.
  • TABLE 3 Apoptosis inducing activity among a series of compounds Fold induction at 100 ⁇ M Reference compounds Indomethacin ⁇ 2.0 MY5445 4.7 Sulindac sulfide 7.9 Exisulind ⁇ 2.0 Test compounds A ⁇ 2.0 B 3.4 C 5.6 D ⁇ 2.0 E 4.6
  • apoptosis inducing activity for a series of phosphodiesterase inhibitors was determined. The data are shown in Table 4 below.
  • HT-29 cell were treated for 6 days with various inhibitors of phospohodiesterase.
  • Apoptosis and necrosis were determined morphologically after acridine orange and ethidium bromide labelling in accordance with the assay of section 4.A., supra.
  • the data show that PDE5 is useful for screening compounds that induce apoptosis of HT-29 cells.
  • FIG. 11 shows the inhibitory effect of various concentrations of sulindac sulfide and exisulind on the growth of HT-29 cells.
  • HT-29 cells were treated for six days with various doses of exisulind (triangles) or sulfide (squares) as indicated.
  • Cell number was measured by a sulforhodamine assay as previously described (Piazza et al., Cancer Research, 55: 3110-3116, 1995).
  • the IC 50 value for the sulfide was approximately 45 ⁇ M and 200 ⁇ M for the sulfone.
  • the data shows that both sulindac sulfide and exisulind are capable of inhibiting tumor cell growth.
  • FIG. 12 shows the growth inhibitory and apoptosis-inducing activity of sulindac sulfide.
  • a time course experiment is shown involving HT-29 cells treated with either vehicle, 0.1% DMSO (open symbols) or sulindac sulfide, 120 ⁇ M (closed symbols).
  • Growth inhibition top was measured by counting viable cells after trypan blue staining.
  • Apoptosis bottom was measured by morphological determination following staining with acridine orange and ethidium bromide as described previously (Duke and Cohen, In: Current Protocols in Immunology, 3.17.1-3.17.16, New York, John Wiley and Sons. 1992).
  • the data demonstrate that sulindac sulfide is capable of inhibiting tumor cell growth and that the effect is accompanied by an increase In apoptosis. All data were collected from the same experiment.
  • FIG. 13 shows the growth inhibitory activity of test compound E.
  • HT-29 colon adenocarcinoma cells were treated with the indicated concentration of compound E for six days and cell number was determined by the SRB assay. The calculated IC 50 value was 0.04 ⁇ M.
  • the growth inhibitory activity for a series of phosphodiesterase inhibitors was determined. The data are shown in Table 6 below. HT-29 cell were treated for 6 days with various inhibitors of phospohodiesterase. Cell growth was determined by the SRB assay in accordance with section 3.A., supra. The data show that inhibitors of PDE5 were effective for inhibiting tumor cell growth.
  • FIG. 14 shows the inhibition of premalignant lesions in mammary gland organ culture by sulindac metabolites.
  • Mammary gland organ culture experiment were performed as previously described (Mehta and Moon, Cancer Research, 46: 5832-5835, 1986). The results demonstrate that sulindac and exisulind effectively inhibit the formation of premalignant lesions, while sulindac sulfide was inactive. The data support the hypothesis that cyclooxygenase inhibition is not necessary for the anti-neoplastic properties of desired compounds.
  • test compounds can be ranked according to their potential use for treating neoplasia in humans. Those compounds having desirable effects may be selected for more expensive and time consuming animal studies that are required to get approval before initiating human clinical trials.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

This invention provides a method to identify compounds potentially useful for the treatment of neoplasia in mammals. The phosphodiesterase inhibitory activity of a compound is determined along with COX inhibitory activity. Growth inhibitory and apoptosis inducing effects on cultured tumor cells are also determined. Compounds that exhibit phosphodiesterase inhibiton, growth inhibition and apoptosis induction, but not substantial prostaglandin inhibitory activity, are desirable for the treatment of neoplasia.

Description

    BACKGROUND OF THE INVENTION
  • This invention provides a method for identifying compounds potentially useful for the treatment and prevention of pre-cancerous and cancerous lesions in mammals. This application is a continuation-in-part of U.S. patent application Ser. No. 08/866,027 to Piazza et al. Filed May 30, 1997. [0001]
  • Familial adenomatous polyposis (“FAP”) is an inherited disease where the victim's colon contains many polyps or adenomas virtually uncountable in most instances. Because such patients develop so many polyps or adenomas each of which has a significant risk of developing into a cancer—the typical treatment is surgical removal of the colon. In about 1983, Waddell discovered that the nonsteroidal anti-inflammatory drug (“NSAID”) sulindac would cause colonic polyps (a type of pre-cancerous lesion) to regress and prevent their recurrence when that drug was administered to patients with FAP. Waddell's experience with sulindac in FAP patients was confirmed in several subsequent studies. Unfortunately, since sulindac and other NSAIDS aggravate the digestive tract (not to mention side effects involving kidney and interference with normal blood clotting) of patients to whom it has been chronically administered, it is not a practical treatment for FAP or any other cancer or precancerous indication (i.e., neoplasia) requiring long-term administration. [0002]
  • Waddell originally hypothesized that the mechanism of action of sulindac on colonic polyps involved the inhibition of the synthesis of prostaglandin (PG). (Waddell, W. R. et al., “Sulindac for Polyposis of the Colon,” [0003] Journal of Surgical Oncology, 24:83-87, 1983). Prostaglandin (“PG”) synthesis inhibition results from the inhibition of cyclooxygenase (COX) caused by NSAIDs. A common benefit of NSAIDs is the reduction of inflammation, which is known to be caused by the reduction of PG levels. Since NSAIDs are known to inhibit COX, which inhibits PG synthesis, it is widely believed that the regression of colonic polyps is attributed to this property. In fact, notwithstanding recent discoveries to the contrary, it has become conventional wisdom that administration of an inhibitor of PG synthesis (e.g., an NSAID) to a patient with FAP or other precancerous or cancerous lesion will result in the regression of the lesion due to a reduction of PG levels.
  • Recent discoveries, however, are leading scientists in a completely different direction—that it is not necessary to inhibit COX to treat neoplasia patients successfully. Pamukcu et al., in U.S. Pat. No. 5,401,774, disclosed that sulfonyl compounds, that were previously reported to be inactive as PG synthesis inhibitors (and therefore not an NSAID or an anti-inflammatory compound) unexpectedly inhibited the growth of a variety of neoplastic cells, including colon polyp cells. These sulfonyl derivatives have proven effective in rat models of colon carcinogenesis, and one variant (now referred to as exisulind) has proven effective in preliminary human clinical trials with FAP patients. [0004]
  • The importance of this discovery—and the de-linking of anti-neoplasitic activity and COX inhibition—cannot be overstated. If those two phenomena were related, there would be little hope for a safe NSAID therapy for FAP patients because the side effects of NSAIDs, such as gastric irritation, are also caused by COX inhibition. Prostaglandins play a protective function in the lining of the stomach. When NSAIDs are administered, COX is inhibited and PG levels are reduced: gastric irritation is a common result. Those side effects may not manifest themselves in short-term (acute) NSAID therapy. However, during long-term (chronic) NSAID therapy, gastric irritation, bleeding and ulceration are very common. In significant numbers of cases, NSAID therapy must be stopped due to the severity of those side effects and other potentially lethal side effects. Furthermore, the severity of such side effects increases with age, probably because natural PG levels in gastric mucosa falls with age. Thus, useful compounds for treating neoplastic lesions should desirably inhibit neoplastic cell growth, but should not inhibit COX. [0005]
  • Conventional methods for screening compounds may be used to find improved compounds that inhibit neoplastic cell growth. Under this scenario, drugs may be screened using in vitro models. But conventional in vitro screening methods could pass many compounds that later are shown to be ineffective in animal models because of a number of unanticipated problems, one of which may be that the in vitro screen is not predictive of efficacy. Animal model studies are time consuming and expensive. Therefore, a more precise in vitro screening method that provides predictive information for treating neoplasia is needed to screen compounds prior to human testing. Knowledge of a specific target for inhibiting human cancer would allow for greater precision and efficiency whereby highly effective and safe compounds can be identified prior to animal testing. [0006]
  • Presently, rational drug discovery methods are being applied in the pharmaceutical industry to improve methods for identifying clinically useful compounds. Typically, rational drug discovery methods relate to a “lock and key” concept whereby structural relationships between a therapeutic target molecule (lock) and pharmaceutical compounds (key) are defined. Such methods are greatly enhanced by specialty computer software that accesses databases of compounds to identify likely geometric fits with the target molecule. Unfortunately, to use these systems, one has to have insight to the target molecule (lock). The target may be an enzyme, a protein, a membrane or nuclear receptor, or a nucleic acid sequence, for example. [0007]
  • In complex diseases, such as neoplasia, scientists have identified a number of potential targets. However, many of the drugs available for the treatment of neoplasia are non-specific and toxic to normal tissues, and are not indicated for precancer and used only when neoplastic cells progress to cancer. Greater understanding of the mechanisms involved in cancer may lead scientists on the path towards designing more specific antineoplastic drugs—drugs that can safely be administered earlier in the disease process. [0008]
  • SUMMARY OF THE INVENTION
  • This invention relates to a novel in vitro method for screening test compounds for their ability to treat and prevent neoplasia, especially pre-cancerous lesions, safely. In particular, the present invention provides a method for identifying test compounds that can be used to treat and prevent neoplasia, including precancerous lesions, with minimal side effects associated with COX inhibition and other non-specific interactions. [0009]
  • In one embodiment of this invention, therefore, the screening method involves determining the COX inhibition activity of a test compound. Because the inventors have discovered a relationship between inhibition of cancer and inhibition of phosphodiesterase Type-5 isoenzyme (“PDE5”), this invention includes determining the PDE5 inhibition activity of the compound. Preferably, the screening method of this invention farther includes determining whether the compounds inhibit the growth of tumor cells in a cell culture. [0010]
  • In an alternate embodiment, the screening method of this invention involves determining the COX inhibition activity of the compound, determining the PDE5 inhibition activity of the compound and determining whether the compound induces apoptosis in tumor cells. [0011]
  • By screening compounds in this fashion, potentially beneficial and improved compounds can be identified more rapidly and with greater precision than possible in the past. Further benefits will be apparent from the detailed description that follows.[0012]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates the effect of the sulfide derivative of sulindac and the sulfone derivative of sulindac (a.k.a. exisulind) on purified cyclooxygenase activity. [0013]
  • FIG. 2 illustrates the effects of test compounds B and E on COX inhibition. [0014]
  • FIG. 3 illustrates the inhibitory effects of sulindac sulfide and exisulind on PDE-4 and PDE5 purified from cultured tumor cells. [0015]
  • FIG. 4 illustrates the effects of sulindac sulfide on cyclic nucleotide levels in HT-29 cells. [0016]
  • FIG. 5 illustrates the phosphodiesterase inhibitory activity of compound B. [0017]
  • FIG. 6 illustrates the phosphodiesterase inhibitory activity of compound E. [0018]
  • FIG. 7 illustrates the effects of sulindac sulfide and exisulind on apoptosis and necrosis of HT-29 cells. [0019]
  • FIG. 8 illustrates the effects of sulindac sulfide and exisulind on HT-29 cell growth inhibition and apoptosis induction as determined by DNA fragmentation. [0020]
  • FIG. 9 illustrates the apoptosis inducing properties of compound E. [0021]
  • FIG. 10 illustrates the apoptosis inducing properties of compound B. [0022]
  • FIG. 11 illustrates the effects of sulindac sulfide and exisulind on tumor cell growth. [0023]
  • FIG. 12 illustrates the growth inhibitory and apoptosis-inducing activity of sulindac sulfide and control (DMSO). [0024]
  • FIG. 13 illustrates the growth inhibitory activity of compound E. [0025]
  • FIG. 14 illustrates the inhibition of pre-malignant, neoplastic lesions in mouse mammary gland organ culture by sulindac metabolites. [0026]
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The method of this invention is useful to identify compounds that can be used to treat or prevent neoplasms, and which are not characterized by the serious side effects of conventional NSAIDs. [0027]
  • Cancer and precancer may be thought of as diseases that involve unregulated cell growth. Cell growth involves a number of different factors. One factor is how rapidly cells proliferate, and another involves how rapidly cells die. Cells can die either by necrosis or apoptosis depending on the type of environmental stimuli. Cell differentiation is yet another factor that influences tumor growth kinetics. Resolving which of the many aspects of cell growth is affected by a test compound is important to the discovery of a relevant target for pharmaceutical therapy. Screening assays based on this selectivity can be combined with tests to determine which compounds having growth inhibiting activity. [0028]
  • This invention is the product of several important discoveries. First, the present inventors discovered that desirable inhibitors of tumor cell growth induce premature death of cancer cells by apoptosis (see, Piazza, G. A., et al., [0029] Cancer Research, 55(14), 3110-16, 1995). Second, the present inventors unexpectedly discovered that compounds that selectively induce apoptosis without substantial COX inhibition also inhibit phosphodiesterase (“PDE”). In particular, and contrary to leading scientific studies, desirable compounds for treating neoplastic lesions selectively inhibit Type 5 isoenzyme form of phosphodiesterase (“PDE5”) (EC 3.1.4.17). PDE5 is one of at least seven isoenzymes of phosphodiesterase. PDE5 is unique in that it selectively degrades cyclic GMP, while the other types of PDE are either non-selective or degrade cyclic AMP. Preferably, desirable compounds do not substantially inhibit other phosphodiesterase types.
  • A preferred embodiment of the present invention involves determining the cyclooxygenase inhibition activity of a given compound, and determining the PDE5 inhibition activity of the compound. The test compounds are scored for their probable ability to treat neoplastic lesions either directly by assessing their activities against specific cutoff values or indirectly by comparing their activities against known compounds useful for treating neoplastic lesions. A standard compound that is known to be effective for treating neoplastic lesions without causing gastric iritation is 5-fluoro-2-methyl-1-(p-methylsulfonylbenzylidene)-3-indenylacetic acid (“exisulind”). Other useful compounds for comparative purposes include those that are known to inhibit COX, such as indomethacin and the sulfide metabolite of sulindac: 5-fluoro-2-methyl-1-(p-methylsulfinylbenzylidene)-3-indenylacetic acid (“sulindac sulfide”). Other useful compounds for comparative purposes include those that are known to inhibit PDE5, such as 1-(3-chloroanilino)-4-phenyphthalazine (“MY5445”). [0030]
  • A test compound is clearly determined to be a promising candidate if it performs better than or comparable to exisulind and does not inhibit COX. In general, desirable compounds are those that inhibit PDE5 and inhibit cell growth and induce apoptosis, but do not inhibit COX at pharmacologically accepted doses. [0031]
  • As used herein, the term “precancerous lesion” includes syndromes represented by abnormal neoplastic, including dysplastic, changes of tissue. Examples include dysplastic growths in colonic, breast, prostate or lung tissues, or conditions such as dysplastic nevus syndrome, a precursor to malignant melanoma of the skin. Examples also include, in addition to dysplastic nevus syndromes, polyposis syndromes, colonic polyps, precancerous lesions of the cervix (i.e., cervical dysplasia), esophagus, lung, prostatic dysplasia, prostatic intraneoplasia, breast and/or skin and related conditions (e.g., actinic keraosis), whether the lesions are clinically identifiable or not. [0032]
  • As used herein, the term “carcinoma” or “cancer” refers to lesions which are cancerous. Examples include malignant melanomas, breast cancer, prostate cancer and colon cancer. As used herein, the terms “neoplasia” and “neoplasms” refer to both cancerous and pre-cancerous lesions. [0033]
  • As used herein, the abbreviation PG represents prostaglandin; PS represents prostaglandin synthetase; PGE[0034] 2 represents prostaglandin E2; PDE represents phosphodiesterase; COX represents cyclooxygenase; RIA represents—radioimmunoassay.
  • As used herein, “PDE5” refers to that enzyme and any of its isoforms that exhibit cGMP specific hydrolytic enzyme activities and high affinity cGMP binding. [0035]
  • In another aspect of the invention, there is a method for treating patients in need of treatment for neoplasia by identifying compounds that exhibit substantial PDE5 inhibitory activity at pharmacologically acceptable doses, and administering one or more of those compounds to a patient in need thereof with neoplasia sensitive to the compound. [0036]
  • SCREENING PROTOCOLS
  • The following screening protocols, and alternative protocols, are provided to aid in the understanding of the preferred methods used to screen test compounds to determine their potential to treat or prevent neoplasia, especially pre-cancerous lesions. [0037]
  • 1. Determining COX Inhibitory Activity [0038]
  • COX inhibition can be determined by either of two methods. One method involves measuring PGE[0039] 2 secretion by intact HL-60 cells following exposure to the compound being screened. The other method involves measuring the activity of purified cyclooxygenases (COXs) in the presence of the compound. Both methods involve protocols previously described in the literature.
  • 1.A. PGE[0040] 2 Secretion
  • Compounds of this can be evaluated to determine whether they inhibited the production of prostaglandin E[0041] 2 (“PGE2”), according to procedures known in the art. For example, PGE2 secreted from a cell can be measured using an enzyme immunoassay (EIA) kit for PGE2, such as commercially available from Amersham, Arlington Heights, Ill. USA. Suitable cells include those which make an abundance of PG, such as HL-60 cells. HL-60 cells are human promyelocytes that are differentiated with DMSO in mature granulocytes. (See, Collins, S. J., Ruscetti, F. W., Gallagher, R. E. and Gallo, R. C., “Normal Functional Characteristics of Cultured Human Promyelocytic Leukemia Cells (HL-60) After Induction of Differentiation By Dimethylsulfoxide”, J. Exp. Med., 149:969-974, 1979). These differentiated cells produce PGE2 after stimulation with a calcium ionophore A23187 (see, Kargman, S., Prasit, P. and Evans, J. F., “Translocation of HL-60 Cell 5-Lipoxygenase”, J. Biol. Chem., 266: 23745-23752, 1991). HL-60 are available from the American Type Culture Collection (ATCC:CCL240). They can be crown in a RPMI 1640 medium supplemented with 20% heat-inactivated fetal bovine serum, 50 U/ml penicillin and 50 μg/ml streptomycin in an atmosphere of 5% CO2 at 37° C. To induce myeloid differentiation, cells are exposed to 1.3% DMSO for 9 days and then washed and resuspended in Dulbecco's phosphate-buffered saline at 3×106 cells/ml.
  • The differentiated HL-60 cells (3×10[0042] 6 cells/ml) can be incubated for 15 min at 37° C. in the presence of the compounds tested at the desired concentration. Cells are then stimulated by A23187 (5×10−6M) for 15 min. PGE2 secreted into the external medium is measured as described above.
  • 1.B Purified Cyclooxygenases [0043]
  • Two different forms of cyclooxygenase (COX-I and COX-2) have been reported in the literature to regulate prostaglandin synthesis. It is known that COX-2 represents the inducible form of COX while COX-I represents a constitutive form. COX-I activity can be measured using the method described by Mitchell et al. (“Selectivity of Nonsteroidal Anti-inflammatory Drugs as Inhibitors of Constitutive and Inducible Cyclooxygenase,” [0044] Proc. Natl. Acad. Sci. USA., 90:11693-11697, 1993, which is incorporated herein by reference) using COX-I purified from ram seminal vesicles as described by Boopathy & Balasubramanian, “Purification And Characterization Of Sheep Platelet Cyclooxygenase” (Biochem. J., 239:371-377, 1988, which is incorporated herein by reference). COX-2 activity can be measured using COX-2 purified from sheep placenta as described by Mitchell et al., 1993, supra.
  • The cyclooxygenase inhibitory activity of a drug can be determined by methods known in the art. For example, Boopathy & Balasubramanian, 1988, supra, described a procedure in which prostaglandin H synthase 1 (Cayman Chemical, Ann Arbor, Mich.) is incubated at 37° C. for 20 min with 100 μM arachidonic acid (Sigma Chemical Co.), cofactors (such as 1.0 mM glutathione, 1.0 mM hydroquinone, 0.625 μM hemoglobin and 1.25 mM CaCl[0045] 2 in 100 mM Tris-HCl, pH 7.4) and the drug to be tested. Following incubation, the reaction can be terminated with trichloroacetic acid. Enzymatic activity can then be measured spectrophotometrically at 530 nm after stopping the reaction by adding thiobarbituric acid and malonaldehyde.
  • Obviously, a compound that exhibits minimal COX-I or COX-2 inhibitory activity in relation to its greater PDE5 inhibitory activity may not be entirely undesirable. [0046]
  • 1.C. Analyzing Results [0047]
  • The amount of inhibition is determined by comparing the activity of the cyclooxygenase in the presence and absence of the test compound. Residual or no COX inhibitory activity (i.e., less than about 25%) at a concentration of about 100 μM is indicative that the compound should be evaluated further for usefulness for treating neoplasia. Preferably, the IC[0048] 50 concentration should be greater than 1000 μM for the compound to be further considered potential use.
  • 2. Determining Phosphodiesterase (PDE5) Inhibition Activity [0049]
  • Compounds can be screened for inhibitory effect on phosphodiesterase activity using either the enzyme isolated from any tumor cell line such as HT-29 or SW-480, or recombinant HS-PDE5, for example, or measuring cyclic nucleotide levels in whole cells. [0050]
  • 2.A. Enzyme Assay [0051]
  • Phosphodiesterase activity can be determined using methods known in the art, such as a method using radioactive [0052] 3H cyclic GMP (cGMP)(cyclic 3′,5′-guanosine monophosphate) as the substrate for PDE5 enzyme. (Thompson, W. J., Teraski, W. L., Epstein, P. M., Strada, S. J., Advances in Cyclic Nucleotide Research, 10:69-92, 1979, which is incorporated herein by reference). In brief, a solution of defined substrate 3H-cGMP specific activity (0.2 μM; 100,000 cpm; containing 40 mM Tris-HCl (pH 8.0), 5 mM MgCl2 and 1 mg/ml BSA) is mixed with the drug to be tested in a total volume of 400 μl. The mixture is incubated at 30° C. for 10 minutes with partially purified PDE5 isolated from HT-29 cells. Reactions are terminated, for example, by boiling the reaction mixture for 75 seconds. After cooling on ice, 100 μl of 0.5 mg/ml snake venom (O. Hannah venom available from Sigma) is added and incubated for 10 min at 30° C. This reaction is then terminated by the addition of an alcohol, e.g. 1 ml of 100% methanol. Assay samples are applied to a anion chromatography column (1 ml Dowex, from Aldrich) and washed with 1 ml of 100% methanol. The amount of radioactivity in the breakthrough and the wash from the columns in then measured with a scintillation counter. The degree of PDE5 inhibition is determined by calculating the amount of radioactivity in drug-treated reactions and comparing against a control sample (a reaction mixture lacking the tested compound).
  • 2.B. Cyclic Nucleotide Measurements [0053]
  • Alternatively, the ability for desirable compounds to inhibit PDE5 is reflected by an increase in cGMP in neoplastic cells exposed to a compound being screened. The amount of PDE5 activity can be determined by assaying for the amount of cyclic GMP in the extract of treated cells using radioimmunoassay (RIA). In this procedure, HT-29 or SW-480 cells are plated and grown to confluency. The test compound is then incubated with the cell culture at a concentration of compound between about 200 μM to about 200 pM. About 24 to 48 hours thereafter, the culture media is removed from the cells, and the cells are solubilized. The reaction is stopped by using 0.2N HCl/50%-MeOH. A sample is removed for protein assay. Cyclic GMP is purified from the acid/alcohol extracts of cells using anion-exchange chromatography, such as a Dowex column. The cGMP is dried, acetylated according to published procedures, such as using acetic anhydride in triethylamine, (Steiner, A. L., Parker, C. W., Kipnis, D. M., [0054] J. Biol Chem., 247(4): 1106-13, 1971, which is incorporated herein by reference). The acetylated cGMP is quantitated using radioimmunoassay procedures (Harper, J., Brooker, G., Advances in Nucleotide Research, 10:1-33, 1979, which is incorporated herein by reference). Iodinated ligands (tyrosine metheyl ester) of derivatized cyclic GMP are incubated with standards or unknowns in the presence of antisera and appropriate buffers. Antiserum may be produced using cyclic nucleotide-haptene directed techniques. The antiserum is from sheep injected with succinyl-cGMP-albumin conjugates and diluted 1/20,000. Dose-interpolation and error analysis from standard curves are applied as described previously (Seibert, A. F., Thompson, W. J., Taylor, A., Wilbourn, W. H., Barnard, J. and Haynes, J., J. Applied Physio., 79:389-395, 1992, which is incorporated hereinby reference).
  • In addition, the culture media may be acidified, frozen (−70° C.) and also analyzed for cGMP and cAMP. [0055]
  • In addition to observing increases in content of cGMP caused by desirable test compounds, decreases in content of cAMP have been observed. It has been observed that a particularly desirable compound (i.e. one that selectively induces apoptosis in neoplastic cells, but not substantially in normal cells) follows a time course consistent with PDE5 inhibition as one initial action resulting in an increased cGMP content within minutes. Secondarily, treatment of neoplastic cells with a desirable anti-neoplastic compound leads to decreased cAtIP content within 24 hours. The intracellular tarcets of drug actions are being studied further, but current data supports the concept that both the initial rise in cGMP content followed by the subsequent fall in cAMP content precede apoptosis in neoplastic cells exposed to desirable compounds. [0056]
  • The change in the ratio of the two cyclic nucleotides may be a more accurate tool for evaluating desirable PDE5 inhibition activity of test compounds, rather than measuring only the absolute value of cGMP, only PDE5 inhibition, or only the absolute value of cGMP. In neoplastic cells not treated with anti-neoplastic compounds, the ratio of cGMP content/cAMNP content is in the 0.03-0.05 range (i.e., 300-500 fmol/mg protein cGMP content over 6000-8000 fmol/mg protein cAMP content). After exposure to desirable anti-neoplastic compounds, that ratio increases several fold (preferably at least about a three-fold increase) as the result of an initial increase in cyclic GMP and the later decrease in cyclic AMP. [0057]
  • Specifically, it has been observed that particularly desirable compounds achieve an initial increase in cGMP content in treated neoplastic cells to a level of cGMP greater than about 500 fmol/mg protein. In addition, particularly desirable compounds cause the later decrease in cAMP content in treated neoplastic cells to a level of cAMP less than about 4000 fmol/mg protein. [0058]
  • To determine the content of cyclic AMP, radioimmunoassay techniques similar to those described above for cGMP are used. Basically, cyclic nucleotides are purified from acid/alcohol extracts of cells using anion-exchange chromatography, dried, acetylated according to published procedures and quantitated using radioimmunoassay procedures. Iodinated ligands of derivatized cyclic AMP and cyclic GMP are incubated with standards or unknowns in the presence of specific antisera and appropriate buffers. [0059]
  • Verification of the cyclic nucleotide content may be obtained by determining the turnover or accumulation of cyclic nucleotides in intact cells. To measure inteact cell cAMP, [0060] 3H-adenine prelabeling is used according to published procedures (Whalin M. E., R. L. Garrett Jr., W. J. Thompson, and S. J. Strada, “Correlation of cell-free brain cyclic nucleotide phosphodiesterase activities to cyclic AMP decay in intact brain slices”, Sec. Mess. and Phos. Protein Research, 12:31 1-325, 1989, which is incorporated herein by reference). The procedure measures flux of labeled ATP to cyclic AMP and can be used to estimate intact cell adenylate cyclase or cyclic nucleotide phosphodiesterase activities depending upon the specific protocol. Cyclic GMP accumulation was too low to be studied with intact cell prelabeling according to published procedures (Reynolds, P. E., S. J. Strada and W. J. Thompson, “Cyclic GMP accumulation in pulmonary microvascular endothelial cells measured by intact cell prelabeling,” Life Sci., 60:909-918, 1997, which is incorporated herein by reference).
  • 2.C. Tissue Sample Assay [0061]
  • The PDE5 inhibitory activity of a test compound can also be determined from a tissue sample. Tissue samples, such as mammalian (preferably rat) liver, are collected from subjects exposed to the test compound. Briefly, a sample of tissue is homogenized in 500 μl of 6% TCA. A known amount of the homogenate is removed for protein analysis. The remaining homogenate is allowed to sit on ice for 20 minutes to allow for the protein to precipitate. Next, the homogenate is centrifuged for 30 minutes at 15,000 g at 4° C. The supernatant is recovered and the pellet recovered. The supernatant is washed four times with five volumes of water saturated diethyl ether. The upper ether layer is discarded between each wash. The aqueous ether extract is dried in a speed vac. Once dried, the sample can be frozen for future use, or used immediately. The dried extract is dissolved in 500 μl of assay buffer. The amount of PDE5 inhibition is determined by assaying for the amount of cyclic nucleotides using an enzyme immunoassay (EIA), such as the Biotrak EIA system acetylation protocol (available from Amersham, Arlington Heights, Ill., USA). Alternatively, RIA procedures as detailed above may be used. [0062]
  • 2.D. Analyzing Results [0063]
  • The amount of inhibition is determined by comparing the activity of PDE5 in the presence and absence of the test compound. Inhibition of PDE5 activity is indicative that the compound is useful for treating neoplasia. Significant inhibitory activity greater than that of the benchmark, exisulind, preferably greater than 50% at a concentration of 10 μM or below, is indicative that a compound should be further evaluated for antineoplastic properties. Preferably, the IC[0064] 50 value for PDE5 inhibition should be less than 50 μM for the compound to be further considered for potential use.
  • 3. Determining Whether a Compound Reduces the Number of Tumor Cells [0065]
  • In an alternate embodiment, the screening method of the present invention involves further determining whether the compound reduces the growth of tumor cells. Various cell lines can be used in the sample depending on the tissue to be tested. For example, these cell lines include: SW-480—colonic adenocarcinoma; HT-29—colonic adenocarcinoma, A-427—lung adenocarcinoma carcinoma; MCF-7—breast adenocarcinoma; and UACC-375—melanoma line; and DU145—prostrate carcinoma. Cytotoxicity data obtained using these cell lines are indicative of an inhibitory effect on neoplastic lesions. These cell lines are well characterized. and are used by the United States National Cancer Institute in their screening program for new anti-cancer drugs. [0066]
  • 3A. Tumor Inhibition in HT-29 Cell Line [0067]
  • A compound's ability to inhibit tumor cell growth can be measured using the HT-29 human colon carcinoma cell line obtained from ATCC (Bethesda, Md.). HT-29 cells have previously been characterized as relevant colon tumor cell culture model (Fogh, J., and Trempe, G. [0068] In: Human Tumor Cells in Vitro, J. Fogh (eds.), Plenum Press, New York, pp. 115-159, 1975). HT-29 cells are maintained in RPMI media supplemented with 5% fetal serum (Gemini Bioproducts, Inc., Carlsbad, Calif.) and 2 mm glutamine, and 1% antibiotic-antimycotic in a humidified atmosphere of 95% air and 5% CO2 at 37° C. Briefly, HT-29 cells are plated at a density of 500 cells/well in 96 well microtiter plates and incubated for 24 hours at 37° C. prior to the addition of compound. Each determination of cell number involved six replicates. After six days in culture, the cells are fixed by the addition of cold trichloroacetic acid to a final concentration of 10% and protein levels are measured using the sulforhodamine B (SRB) colorimetric protein stain assay as previously described by Skehan, P., Storeng, R., Scudiero, D., Monks, A., McMahon, J., Vistica, D., Warren, J. T., Bokesch, H., Kenney, S., and Boyd, M. R., “New Colorimetric Assay For Anticancer-Drug Screening,” J. Natl. Cancer Inst. 82: 1107-1112, 1990, which is incorporated herein by reference.
  • In addition to the SRB assay, a number of other methods are available to measure growth inhibition and could be substituted for the SRB assay. These methods include counting viable cells following trypan blue staining, labeling cells capable of DNA synthesis with BrdU or radiolabeled thymidine, neutral red staining of viable cells, or MTT staining of viable cells. [0069]
  • 3.B. Analyzing Results [0070]
  • Significant tumor cell growth inhibition greater than about 50% at a dose of 100 μM or below is further indicative that the compound is useful for treating neoplastic lesions. Preferably, an IC[0071] 50 value is determined and used for comparative purposes. This value is equivalent to the concentration of drug needed to inhibit tumor cell growth by 50% relative to the control. Preferably, the IC50 value should be less than 100 μM for the compound to be considered further for potential use for treating neoplastic lesions.
  • 4. Determining Whether a Compound Induces Apoptosis [0072]
  • In a second alternate embodiment, the screening method of the present invention further involves determining whether the compound induces apoptosis in cultures of tumor cells. [0073]
  • Two distinct forms of cell death may be described by morphological and biochemical criteria: necrosis and apoptosis. Necrosis is accompanied by increased permeability of the plasma membrane; the cells swell and the plasma membrane ruptures within minutes. Apoptosis is characterized by membrane blebbing, condensation of cytoplasm and the activation of endogenous endonucleases. [0074]
  • Of the two, apoptosis is the most common form of eukaryotic cell death. It occurs naturally during normal tissue turnover and during embryonic development of organs and limbs. Apoptosis also is induced by cytotoxic T-lymphocytes and natural killer cells, by ionizing radiation and certain chemotherapeutic drugs. Inappropriate regulation of apoptosis is thought to play an important role in many pathological conditions including cancer, AIDS, Alzheimer disease, etc. Compounds can be screened for induction of apoptosis using cultures of tumor cells maintained under conditions as described above. Treatment of cells with test compounds involves either pre- or post-confluent cultures and treatment for two to seven days at various concentrations. Apoptotic cells are measured in both the attached and “floating” compartments of the cultures. Both compartments are collected by removing the supernatant, trypsinizing the attached cells, and combining both preparations following a centrifugation wash step (10 minutes, 2000 rpm). The protocol for treating tumor cell cultures with sulindac and related compounds to obtain a significant amount of apoptosis has been described in the literature. (See, Piazza, G. A., et al., [0075] Cancer Research, 55:3110-16, 1995, which is incorporated herein by reference). The novel features include collecting both floating and attached cells, identification of the optimal treatment times and dose range for observing apoptosis, and identification of optimal cell culture conditions.
  • 4.A. Morphological Observation of Apoptosis [0076]
  • Following treatment with a test compound, cultures can be assayed for apoptosis and necrosis by florescent microscopy following labeling with acridine orange and ethidium bromide. The method for measuring apoptotic cell number has previously been described by Duke & Cohen, “Morphological And Biochemical Assays Of Apoptosis,” [0077] Current Protocols In Immunology, Coligan et al., eds., 3.17.1-3.17.16 (1992, which is incorporated herein by reference).
  • For example, floating and attached cells can be collected by trypsinization and washed three times in PBS. Aliquots of cells can be centrifuged. The pellet can then be resuspended in media and a dye mixture containing acridine orange and ethidium bromide prepared in PBS and mixed gently. The mixture can then be placed on a microscope slide and examined. [0078]
  • 4.B. Analysis of Apoptosis by DNA Fragmentation [0079]
  • Apoptosis can also be quantified by measuring an increase in DNA fragmentation in cells which have been treated with test compounds. Commercial photometric EIA for the quantitative in vitro determination of cytoplasmic histone-associated-DNA-fragments (mono- and oligonucleosomes) are available (Cell Death Detection ELISA[0080] okys, Cat. No. 1,774,425, Boehringer Mannheim). The Boehringer Mannheim assay is based on a sandwich-enzyme-immunoassay principle using mouse monoclonal antibodies directed against DNA and histones, respectively. This allows the specific determination of mono-and oligonucleosomes in the cytoplasmatic fraction of cell lysates.
  • According to the vendor, apoptosis is measured in the following fashion. The sample (cell-lysate) is placed into a streptavidin-coated microtiter plate (“MTP”). Subsequently, a mixture of anti-histone-biotin and anti-DNA peroxidase conjugate are added and incubated for two hours. During the incubation period, the anti-histone antibody binds to the histone-component of the nucleosomes and simultaneously fixes the immunocomplex to the streptavidin-coated MTP via its biotinylation. Additionally, the anti-DNA peroxidase antibody reacts with the DNA component of the nucleosomes. After removal of unbound antibodies by a washing step, the amount of nucleosomes is quantified by the peroxidase retained in the immunocomplex. Peroxidase is determined photometrically with ABTS7 (2,2′-Azido-[3 -ethylbenzthiazolin-suffonate])* as substrate. [0081]
  • For example, SW-480 colon adenocarcinoma cells are plated in a 96-well MTP at a density of 10,000 cells per well. Cells are then treated with test compound, and allowed to incubate for 48 hours at 37° C. After the incubation, the MTP is centrifuged and the supernatant is removed. The cell pellet in each well is then resuspended in lysis buffer for 30 minutes. The lysates are then centrifuged and aliquots of the supernatant (i.e. cytoplasmic fraction) are transferred into streptavidin coated MTP. Care is taken not to shake the lysed pellets (i.e. cell nucleii containing high molecular weight, unfragmented DNA) in the MTP. Samples are then analyzed. [0082]
  • Fold stimulation (FS=OD[0083] max/ODveh), an indicator of apoptotic response, is determined for each compound tested at a given concentration. EC50 values may also be determined by evaluating a series of concentrations of the test compound.
  • 4.C. Analyzing Results [0084]
  • Statistically significant increases of apoptosis (i.e., greater than 2 fold stimulation at a concentration of 100 μM) are further indicative that the compound is useful for treating neoplastic lesions. Preferably, the EC[0085] 50 value for apoptotic activity should be less than 100 μM for the compound to be further considered for potential use for treating neoplastic lesions. EC50 is herein defined as the concentration that causes 50% induction of apoptosis relative to vehicle treatment.
  • 5. Validation—Mammary Gland Organ Culture Model Tests [0086]
  • Test compounds identified by the above methods can be tested for antineoplastic activity by their ability to inhibit the incidence of preneoplastic lesions in a mammary gland organ culture system. This mouse mammary gland organ culture technique has been successfully used by other investigators to study the effects of known antineoplastic agents such as NSAIDs, retinoids, tamoxifen, selenium, and certain natural products, and is useful for validation of the screening method of the present invention. [0087]
  • For example, female BALB/c mice can be treated with a combination of estradiol and progesterone daily, in order to prime the glands to be responsive to hormones in vitro. The animals are sacrificed and thoracic mammary glands are excised aseptically and incubated for ten days in growth media supplemented with insulin, prolactin, hydrocortisone, and aldosterone. DMBA (7,12-dimethylbenz(a)anthracene) is administered to induce the formation of premalignant lesions. Fully developed glands are then deprived of prolactin, hydrocortisone, and aldosterone, resulting in the regression of the glands but not the premalignant lesions. [0088]
  • The test compound is dissolved in DMSO and added to the culture media for the duration of the culture period. At the end of the culture period, the glands were fixed in 10% formalin. stained with alum carmine, and mounted on glass slides. The incidence of forming mammary lesions is the ratio of the glands with mammary lesions and glands without lesions. The incidence of mammary lesions in test compound treated glands is compared with that of the untreated glands. [0089]
  • The extent of the area occupied by the mammary lesions can be quantitated by projecting an image of the gland onto a digitation pad. The area covered by the gland is traced on the pad and considered as 100% of the area. The space covered by each of the unregressed structures is also outlined on the digitization pad and quantitated by the computer. [0090]
  • EXPERIMENTAL SECTION
  • A number of test compounds were examined in the various protocols and screened for potential use in treating neoplasia. The results of these tests are reported below. The test compounds are hereinafter designated by a letter code that corresponds to the following: [0091]
  • A—rac-threo-(E)-1-(N,N′-diethylaminoethanethio)-1-(butan-1′,4′-olido)-[3′,4′:1,2]-6-fluoro-2-methyl-3-(p-methylsulfonylbenzylidene)-indan; [0092]
  • B—(Z)-5-Fluoro-2-methyl-1-(3,4,5-trimethoxybenzylidene)-3-acetic acid; [0093]
  • C—(Z)-5-Fluoro-2-methyl-1-(p-chlorobenzylidene)-3-acetic acid; [0094]
  • D—rac-(E)-1-(butan-1′,4′-olido)-[3′, 4′:1,2]-6-fluoro-2-methyl-3-(p-methylsulfonylbenzylidene)-1S-indanyl-N-acetylcysteine; [0095]
  • E—(Z)-5-Fluoro-2-methyl-1-(3,4,5-trimethoxybenzylidene)-3-indenylacetamide, N-benzyl; [0096]
  • F—(Z)-5-Fluoro-2-methyl-1-(p-methylsulfonylbenzylidene)-3-indenylacetamide, N,N′-dicyclohexyl; [0097]
  • G—ribo-(E)-1-Triazolo-[2′,3′:1″,3″]-1-(butan-1′,4′-olido)-[3′,4′:1,2]-6-methyl-3-(p-methylsulfonylbenzylidene)-indan; and [0098]
  • H—rac-(E)-1-(butan-1′,4-olido)-[3′,4′:1,2]-6-fluoro-2-methyl-3′-(p-methylsulfonylbenzylidene)-1S-indanyl-glutathione). [0099]
  • EXAMPLE 1 COX Inhibition Assay
  • Reference compounds and test compounds were analyzed for their COX inhibitory activity in accordance with the protocol for the COX assay of section 1.B. supra. FIG. 1 shows the effect of various concentrations of either sulindac sulfide or exisulind on purified cyclooxygenase (Type 1) activity. Cyclooxygenase activity was determined using purified cyclooxygenase from ram seminal vesicles as described previously (Mitchell et al, supra). The IC-50 value for sulindac sulfide was calculated to be approximately 1.76 μM, while that for exisulind was greater than 10,000 μM. These data show that sulindac sulfide, but not exisulind, is a COX-I inhibitor. Similar data was obtained for the COX-2 isoenzyme. (Thompson, et al., Journal of the National Cancer Institute, 87: 1259-1260, 1995). [0100]
  • FIG. 2 shows the effect of test compounds B and E on COX inhibition. COX activity was determined as for the compounds shown in FIG. 1. The data show that both test compound B and E do not significantly inhibit COX-I. [0101]
    TABLE 1
    Cyclooxygenase inhibitory activity among a series of compounds
    % Inhibition at 100 μM
    Reference compounds
    Indomethacin 95
    MY5445 94
    Sulindac sulfide 97
    Exisulind <25
    Test compounds
    A <25
    B <25
    C 87
    D <25
    E <25
  • In accordance with the protocol of section 1.B., supra, compounds A through E were evaluated for COX inhibitory activity as reported in Table 1 above. Compound C was found to inhibit COX greater than 25% at a 100 μM dose, and therefore, would not be selected for further screening. [0102]
  • EXAMPLE 2 PDE5 Inhibition Assay
  • Reference compounds and test compounds were analyzed for their PDE5 inhibitory activity in accordance with the protocol for the assay of section 2.A., supra. FIG. 3 shows the effect of various concentrations of sulindac sulfide and exisulind on either PDE-4 or PDE5 activity purified from human colon HT-29 cultured tumor cells, as described previously (W. J. Thompson et al., supra). The IC[0103] 50 value of sulindac sulfide for inhibition of PDE4 was 41 μM, and for inhibition of PDE5 was 17 μM. The IC50 value of exisulind for inhibition of PDE4 was 181 μM, and for inhibition of PDE5 was 56 μM. These data show that both sulindac sulfide and exisulind inhibit phospohodiesterase activity. Both compounds show selectivity for the PDE5 isoenzyme form.
  • FIG. 4 shows the effects of sulindac sulfide on either cGMP or cAMP production as determined on cultured HT-29 cells in accordance with the assay of section 2.B., supra. HT-29 cells were treated with sulindac sulfide for 30 minutes and cGMP or cAMP was measured by conventional radioimmunoassay method. As indicated, sulindac sulfide increased the levels of cGMP by greater than 50% with an EC[0104] 50 value of 7.3 μM (top). Levels of cAMP were unaffected by treatment, although a known PDE4 inhibitor, rolipram, increased cAMP (bottom). The data demonstrate the pharmacological significance of inhibiting PDE5, relative to PDE4.
  • FIG. 5 shows the effect of the indicated dose of test compound B on either PDE5 or PDE4 isozymes of phosphodiesterase. The calculated IC[0105] 50 value for PDE5 was 18 μM and 58 μM for PDE4.
  • FIG. 6 shows the effect of the indicated dose of test compound E on either PDE4 or PDE5. The calculated IC[0106] 50 value was 0.08 μM for PDE5 and greater than 25 μM for PDE4.
    TABLE 2
    PDE5 inhibitory activity among a series of compounds
    % Inhibition at 10 μM
    Reference compounds
    Indomethacin 34
    MY5445 86
    Sulindac sulfide 97
    Exisulind 39
    Test compounds
    A <25
    B <25
    C <25
    D 36
    E 75
  • The above compounds in Table 2 were evaluated for PDE inhibitory activity, as described in the protocol of section 2.A; supra. Of the compounds that did not inhibit COX, only compound E was found to cause greater than 50% inhibition at 10 μM. As noted in FIG. 11, compound B showed inhibition of greater than 50% at a dose of 20 μM. Therefore, depending on the dosage level used in a single dose test, some compounds may be screened out that otherwise may be active at slightly higher dosages. The dosage used is subjective and may be lowered after active compounds are found at certain levels to identify even more potent compounds. [0107]
  • EXAMPLE 3 Apoptosis Assay
  • Reference compounds and test compounds were analyzed for their PDE5 inhibitory activity in accordance with the protocol for the assay of section 4.A. and 4.B., supra. In accordance with the assay of 4.A., FIG. 7 shows the effects of sulindac sulfide and exisulind on apoptotic and necrotic cell death. HT-29 cells were treated for six days with the indicated dose of either sulindac sulfide or exisulind. Apoptotic and necrotic cell death was determined previously (Duke and Cohen, In: Current Protocols in Immunology, 3.17.1-3.17.16, New York, John Wiley and Sons, 1992). The data shows that both sulindac sulfide and exisulind are capable of causing apoptotic cell death without inducing necrosis. All data were collected from the same experiment. [0108]
  • In accordance with the assay of 4.B., FIG. 8 shows the effect of sulindac sulfide and sulfone on tumor growth inhibition and apoptosis induction as determined by DNA fragmentation. Top figure; growth inhibition (open symbols, right axis) and DNA fragmentation (closed symbols, left axis) by exisulind. Bottom figure; growth inhibition (open symbols) and DNA fragmentation (closed symbols) by sulindac sulfide. Growth inhibition was determined by the SRB assay after six days of treatment. DNA fragmentation was determined after 48 hours of treatment. All data was collected from the same experiment. [0109]
  • FIG. 9 shows the apoptosis inducing properties of compound E. HT-29 colon adenocarcinoma cells were treated with the indicated concentration of compound E for 48 hours and apoptosis was determined by the DNA fragmentation assay. The calculated EC[0110] 50 value was 0.05 μM.
  • FIG. 10 shows the apoptosis inducing properties of compound B. HT-29 colon adenocarcinoma cells were treated with the indicated concentration of compound B for 48 hours and apoptosis was determined by the DNA fragmentation assay. The calculated EC[0111] 50 value was approximately 175 μM.
    TABLE 3
    Apoptosis inducing activity among a series of compounds
    Fold induction at 100 μM
    Reference compounds
    Indomethacin <2.0
    MY5445 4.7
    Sulindac sulfide 7.9
    Exisulind <2.0
    Test compounds
    A <2.0
    B 3.4
    C 5.6
    D <2.0
    E 4.6
  • In accordance with the protocol of section 4.B., supra, the compounds A through E were tested for apoptosis inducing, activity, as reported in Table 3 above. Compounds B, C and E showed significant apoptotic inducing activity, graeter than 2.0 fold, at a dosage of 100 μM. Of these three compounds, at this dosage only B and E did not inhibit COX and inhibited PDE5. [0112]
  • The apoptosis inducing activity for a series of phosphodiesterase inhibitors was determined. The data are shown in Table 4 below. HT-29 cell were treated for 6 days with various inhibitors of phospohodiesterase. Apoptosis and necrosis were determined morphologically after acridine orange and ethidium bromide labelling in accordance with the assay of section 4.A., supra. The data show that PDE5 is useful for screening compounds that induce apoptosis of HT-29 cells. [0113]
    TABLE 4
    Apoptosis Inducing Data for PDE Inhibitors
    Inhibitor Reported Selectivity % Apoptosis % Necrosis
    Vehicle
    8 6
    8-methoxy-IBMX PDE1 2 1
    Milrinone PDE3 18 0
    RO-20-1724 PDE4 11 2
    MY5445 PDE5 80 5
    IBMX Non-selective 4 13
  • EXAMPLE 4 Growth Inhibition Assay
  • Reference compounds and test compounds were analyzed for their PDE5 inhibitory activity in accordance with the protocol for the assay of section 3.A., supra. FIG. 11 shows the inhibitory effect of various concentrations of sulindac sulfide and exisulind on the growth of HT-29 cells. HT-29 cells were treated for six days with various doses of exisulind (triangles) or sulfide (squares) as indicated. Cell number was measured by a sulforhodamine assay as previously described (Piazza et al., [0114] Cancer Research, 55: 3110-3116, 1995). The IC50 value for the sulfide was approximately 45 μM and 200 μM for the sulfone. The data shows that both sulindac sulfide and exisulind are capable of inhibiting tumor cell growth.
  • FIG. 12 shows the growth inhibitory and apoptosis-inducing activity of sulindac sulfide. A time course experiment is shown involving HT-29 cells treated with either vehicle, 0.1% DMSO (open symbols) or sulindac sulfide, 120 μM (closed symbols). Growth inhibition (top) was measured by counting viable cells after trypan blue staining. Apoptosis (bottom) was measured by morphological determination following staining with acridine orange and ethidium bromide as described previously (Duke and Cohen, In: Current Protocols in Immunology, 3.17.1-3.17.16, New York, John Wiley and Sons. 1992). The data demonstrate that sulindac sulfide is capable of inhibiting tumor cell growth and that the effect is accompanied by an increase In apoptosis. All data were collected from the same experiment. [0115]
  • FIG. 13 shows the growth inhibitory activity of test compound E. HT-29 colon adenocarcinoma cells were treated with the indicated concentration of compound E for six days and cell number was determined by the SRB assay. The calculated IC[0116] 50 value was 0.04 μM.
    TABLE 5
    Growth inhibitory activity among a series of compounds
    % Inhibition at 100 μM
    Reference compounds
    Indomethacin 75
    MY5445 88
    Sulindac sulfide 88
    Exisulind <50
    Test compounds
    A 68
    B 77
    C 80
    D 78
    E 62
  • In accordance with the screening protocol of section 3.A., supra, compounds A through B were tested for growth inhibitory activity, as reported in Table 5 above. All the test compounds showed activity exceeding the benchmark exisulind at a 100 μM single does rest. [0117]
  • The growth inhibitory activity for a series of phosphodiesterase inhibitors was determined. The data are shown in Table 6 below. HT-29 cell were treated for 6 days with various inhibitors of phospohodiesterase. Cell growth was determined by the SRB assay in accordance with section 3.A., supra. The data show that inhibitors of PDE5 were effective for inhibiting tumor cell growth. [0118]
    TABLE 6
    Growth Inhibitory Data for PDE Inhibitors
    Growth inhibition
    Inhibitor Reported Selectivity (IC50, μM)
    8-methoxy-IBMX PDE1 >200 μM
    Milrinone PDE3 >200 μM
    RO-20-1724 PDE4 >200 μM
    MY5445 PDE5
    5 μM
    IBMX Non-selective >100 μM
  • To show the effectiveness of this screening method on various forms of neoplasia, compounds were tested on numerous cell lines. The effects of sulindac sulfide and exisulind on various cell lines was determined. The data is shown in table 7 below. The IC[0119] 50 values were determined by the SRB assay. The data shows the broad effectiveness of these compounds on a broad range of neoplasia, with effectiveness at comparable dose range. Therefore, compounds identified by this invention should be useful for treating multiple forms of neoplasia.
    TABLE 7
    Growth Inhibitory Data of Various Cell Lines
    Cell Type/ IC50 (μM)
    Tissue specificity Sulindac sulfide Exisulind
    HT-29, Colon 60 120
    HCT116, Colon 45 90
    MCF7/S, Breast 30 90
    UACC375, Melanoma 50 100
    A-427, Lung 90 130
    Bronchial Epithelial Cells (normal) 30 90
    NRK, Kidney (normal) 50 180
    KNRK, Kidney (transformed) 60 240
    Human Prostate Carcinoma PC3 82
  • EXAMPLE 5 Activity in Mammary Gland Organ Culture Model
  • FIG. 14 shows the inhibition of premalignant lesions in mammary gland organ culture by sulindac metabolites. Mammary gland organ culture experiment were performed as previously described (Mehta and Moon, [0120] Cancer Research, 46: 5832-5835, 1986). The results demonstrate that sulindac and exisulind effectively inhibit the formation of premalignant lesions, while sulindac sulfide was inactive. The data support the hypothesis that cyclooxygenase inhibition is not necessary for the anti-neoplastic properties of desired compounds.
  • ANALYSIS
  • To identify compounds that have potential use for treating neoplasia, this invention provides a rationale for comparing experimental data of test compounds from several protocols. Within the framework of this invention, test compounds can be ranked according to their potential use for treating neoplasia in humans. Those compounds having desirable effects may be selected for more expensive and time consuming animal studies that are required to get approval before initiating human clinical trials. [0121]
  • Qualitative data of various test compounds and the several protocols are shown in Table 8 below. The data show that exisulind, compound B and compound E exhibit the appropriate activity to pass the screen of four assays: lack of COX inhibition, PDE inhibition, growth inhibition and apoptosis induction. The activity of these compounds in the mammary gland organ culture validates the effectiveness of this invention. The qualitative valuations of the screening protocols rank compound E best, then compound B and then exisulind. [0122]
    TABLE 8
    Activity Profile of Various Compounds
    Mammary
    COX PDE5 Growth Gland Organ
    Compound Inhibition Inhibition Inhibition Apoptosis Culture
    Exisulind ++ ++ ++ +++
    Sulindac sulfide ++++ +++ +++ +++
    MY5445 ++++ +++ +++ +++ +
    A +++ ++ ++
    B +++ +++ +++ ++
    D ++
    E ++++ ++++ ++++ ++++
    F ++ +
    G +++ ++ +++
    H ++
  • Obviously, numerous modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that within the scope of the appended claims, the invention may be practiced otherwise than as specifically described herein. [0123]

Claims (4)

We claim:
1. A method for identifying a compound with potential for treating neoplasia, comprising:
treating neoplastic cells with the compound to be evaluated at a concentration between about 200 μM to 200 pM;
determining the intracellular amount of cGMP in the treated cells; determining the intracellular amount of cAMP in the treated cells;
wherein about a three-fold or greater increase in the ratio of cGMP/cAMP in the treated cells compared to the ratio of cGMP/cAMP in untreated neoplastic cells is indicative that the compound has potential for treating neoplasia.
2. The method of claim 1 wherein an intracellular amount of cGmp in the treated cells greater than 500 fmol/mg protein is indicative that the compound has potential for treating neoplasia.
3. The method of claim 1 wherein an intracellular amount of cAMP in the treated cells less than 4000 fmol/mg protein is indicative that the compound has potential for treating neoplasia.
4. The method of claim 3 wherein an intracellular amount of cAMP in the treated cells less than 4000 fmol/mg protein is indicative that the compound has potential for treating neoplasia.
US10/040,776 1997-05-30 2002-01-07 Method for identifying compounds for inhibition of neoplastic lesions Abandoned US20030004093A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/040,776 US20030004093A1 (en) 1997-05-30 2002-01-07 Method for identifying compounds for inhibition of neoplastic lesions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/866,027 US5858694A (en) 1997-05-30 1997-05-30 Method for identifying compounds for inhibition of cancerous lesions
US4673998A 1998-03-24 1998-03-24
US66403500A 2000-09-18 2000-09-18
US10/040,776 US20030004093A1 (en) 1997-05-30 2002-01-07 Method for identifying compounds for inhibition of neoplastic lesions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US66403500A Continuation 1997-05-30 2000-09-18

Publications (1)

Publication Number Publication Date
US20030004093A1 true US20030004093A1 (en) 2003-01-02

Family

ID=26724247

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/414,625 Expired - Fee Related US6500610B1 (en) 1997-05-30 1999-10-08 Methods for identifying compounds for inhibiting of neoplastic lesions, and pharmaceutical compositions containing such compounds
US10/040,776 Abandoned US20030004093A1 (en) 1997-05-30 2002-01-07 Method for identifying compounds for inhibition of neoplastic lesions
US10/252,983 Abandoned US20030190686A1 (en) 1997-05-30 2002-09-24 Methods for identifying compounds for inhibition of neoplastic lesions, and pharmacetical compositions containing such compounds

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/414,625 Expired - Fee Related US6500610B1 (en) 1997-05-30 1999-10-08 Methods for identifying compounds for inhibiting of neoplastic lesions, and pharmaceutical compositions containing such compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/252,983 Abandoned US20030190686A1 (en) 1997-05-30 2002-09-24 Methods for identifying compounds for inhibition of neoplastic lesions, and pharmacetical compositions containing such compounds

Country Status (16)

Country Link
US (3) US6500610B1 (en)
EP (2) EP0881300B1 (en)
JP (3) JP3053381B2 (en)
KR (1) KR100304859B1 (en)
AT (1) ATE198771T1 (en)
AU (1) AU709666B2 (en)
CA (1) CA2238283C (en)
CZ (1) CZ295868B6 (en)
DE (2) DE69800488T2 (en)
DK (1) DK0881300T3 (en)
ES (1) ES2132055T3 (en)
HK (1) HK1012196A1 (en)
IL (1) IL124699A (en)
NO (1) NO321717B1 (en)
TR (1) TR199800960A2 (en)
TW (1) TW591111B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8271303B2 (en) 2003-09-04 2012-09-18 Hartford Fire Insurance Company System for reducing the risk associated with an insured building structure through the incorporation of selected technologies

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL132366A0 (en) * 1998-10-15 2001-03-19 Cell Pathways Inc Methods for identifying compounds for inhibition of neoplastic lesions and pharmaceutical compositions containing such compounds
US20020009764A1 (en) * 1999-10-08 2002-01-24 W. Joseph Thompson Methods for identifying compounds for inhibition of neoplastic lesions, and pharmaceutical compositions containing such compounds
US20040009464A1 (en) * 1998-10-15 2004-01-15 Li Liu Methods for identifying compounds for inhibition of neoplastic lesions, and pharmacetical compositions containing such compounds
US20050244914A1 (en) * 1999-10-08 2005-11-03 Li Liu Methods for identifying compounds for inhibition of neoplastic lesions, and pharmaceutical compositions containing such compounds
DE10060388A1 (en) * 2000-12-05 2002-06-06 Merck Patent Gmbh Use of pyrazolo [4,3-d] pyrimidines
FR2824334B1 (en) 2001-05-03 2003-10-10 Coletica METHOD FOR TESTING A SUBSTANCE POSSIBLY ACTIVE IN THE FIELD OF LIPOLYSIS AND ITS MAINLY COSMETIC USE
WO2003008578A2 (en) * 2001-07-20 2003-01-30 Board Of Trustees Of The University Of Illinois Reagents and methods for identifying gene targets for treating cancer
DE10135815A1 (en) * 2001-07-23 2003-02-06 Bayer Ag Use of imidazo-triazinone derivative phosphodiesterase 5 inhibitors e.g. for treatment of cardiac insufficiency, psoriasis, diabetes, cancer, glaucoma, bladder disease, Parkinson's disease or pain
DE60237696D1 (en) * 2001-08-01 2010-10-28 Univ Utah AT N-TERMINUS TRUNCATED ISOFORMS OF CYCLIC PHOSPHODIESTERASES PDE3A
WO2003038430A1 (en) * 2001-10-29 2003-05-08 Takeda Chemical Industries, Ltd. Screening method
US20050048573A1 (en) * 2003-02-03 2005-03-03 Plexxikon, Inc. PDE5A crystal structure and uses
WO2005054181A1 (en) * 2003-12-01 2005-06-16 Reverse Proteomics Research Institute Co., Ltd. Novel target protein of anticancer agent and novel anticancer agent (spnal) corresponding thereto
JP2008503446A (en) * 2004-05-06 2008-02-07 プレキシコン,インコーポレーテッド PDE4B inhibitor and use thereof
JP2008512380A (en) * 2004-09-03 2008-04-24 プレキシコン,インコーポレーテッド PDE4B inhibitor
EP1838319B1 (en) * 2005-01-07 2018-03-07 The Johns Hopkins University Pde5 inhibitor compositions and methods for immunotherapy
CA2600822A1 (en) * 2005-03-14 2006-09-21 Nycomed Gmbh Method for preventing cardiovascular diseases
CN101365461B (en) * 2006-01-04 2013-07-24 南方研究所 Derivatives of sulindac, use thereof and preparation thereof
JP2007224007A (en) * 2006-03-27 2007-09-06 Univ De Santiago De Compostela Therapeutic usage of yessotoxin as human tumor cell proliferation suppressor
US8673914B2 (en) 2011-03-28 2014-03-18 St. John's University Use of phosphodiesterase inhibitors for treating multidrug resistance
WO2017168174A1 (en) 2016-04-02 2017-10-05 N4 Pharma Uk Limited New pharmaceutical forms of sildenafil

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4115538A (en) * 1976-02-12 1978-09-19 Yamasu Shoyu Kabushiki Kaisha Assay kit for cAMP and/or cGMP and method of using the same
US5728563A (en) * 1993-01-29 1998-03-17 Taisho Pharmaceutical Co., Ltd. Cyclic nucleotide phosphodiesterase and a method for production thereof
US5760042A (en) * 1985-04-19 1998-06-02 Sankyo Company, Limited Griseolic acid derivatives, their preparation and their use
US5858694A (en) * 1997-05-30 1999-01-12 Cell Pathways, Inc. Method for identifying compounds for inhibition of cancerous lesions
US6156312A (en) * 1993-06-23 2000-12-05 Leskovar; Peter Agents, affecting the hyperactivated immunological effector cells
US6187756B1 (en) * 1996-09-05 2001-02-13 The Massachusetts Institute Of Technology Composition and methods for treatment of neurological disorders and neurodegenerative diseases
US6200771B1 (en) * 1998-10-15 2001-03-13 Cell Pathways, Inc. Method of using a novel phosphodiesterase in pharmaceutical screeing to identify compounds for treatment of neoplasia
US6235782B1 (en) * 1998-11-12 2001-05-22 Rifat Pamukcu Method for treating a patient with neoplasia by treatment with a platinum coordination complex
US6235776B1 (en) * 1998-11-12 2001-05-22 Cell Pathways, Inc. Method for treating a patient with neoplasia by treatment with a paclitaxel derivative

Family Cites Families (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE274218C (en)
GB807826A (en) 1955-03-14 1959-01-21 Thomae Gmbh Dr K Derivatives of pyrimido[5,4-d] pyrimidine and production thereof
US3031450A (en) 1959-04-30 1962-04-24 Thomae Gmbh Dr K Substituted pyrimido-[5, 4-d]-pyrimidines
AT290523B (en) 1962-01-05 1971-06-11 Merck & Co Inc Process for the production of new α- (3-indolyl) -carboxylic acids
US3322755A (en) 1964-03-10 1967-05-30 Boehringer Sohn Ingelheim Basic-substituted 1, 2, 3, 4-tetrahydropyrimido [5, 4-d]-pyrimidines
GB1199768A (en) 1966-10-31 1970-07-22 Pfizer & Co C Nitrogen Heterocycles and process for their preparation
US3812127A (en) 1966-10-31 1974-05-21 Pfizer 4-(quinolin-4-yl)piperazine-1-carboxylic acid esters
US3517005A (en) 1967-10-26 1970-06-23 Pfizer & Co C Certain 2- and 4-substituted quinazolines
US3752826A (en) 1970-01-26 1973-08-14 Mcneilab Inc Aroyl substituted pyrroles
US3654349A (en) 1970-05-01 1972-04-04 Merck & Co Inc Substituted indenyl acetic acids
US3647858A (en) 1970-05-01 1972-03-07 Merck & Co Inc Process for preparing 1-benzylidene-3-indenyl acetic acids
GB1493685A (en) 1970-12-15 1977-11-30 May & Baker Ltd 8-azapurinones
US3819631A (en) 1970-12-15 1974-06-25 May & Baker Ltd Azapurinones
US3780040A (en) 1972-06-02 1973-12-18 R Schnettler 2-substituted-3,4-dihydroquinazolines
JPS536156B2 (en) 1972-10-30 1978-03-04
US4001238A (en) 1976-02-18 1977-01-04 Bristol-Myers Company 1,3,4-oxadiazole amides
US4001237A (en) 1976-02-18 1977-01-04 Bristol-Myers Company Oxazole, isoxazole, thiazole and isothiazole amides
US4060615A (en) 1976-02-18 1977-11-29 Mead Johnson & Company 2-Piperazinyl-6,7-dimethoxyquinazolines
US4076711A (en) 1976-04-05 1978-02-28 Schering Corporation Triazolo [4,5-d]-pyrimidines
US4171363A (en) 1977-02-22 1979-10-16 Bristol-Myers Company 1,2,3-Thiadiazole process
US4101548A (en) 1977-02-22 1978-07-18 Bristol-Myers Company 1,2,3-Thiadiazole amides
US4079057A (en) 1977-05-31 1978-03-14 Bristol-Myers Company Selective immunosuppressive agents
US4098788A (en) 1977-06-20 1978-07-04 Bristol-Myers Company Process for preparing quinazolines
US4102885A (en) 1977-06-20 1978-07-25 Bristol-Myers Company Process for preparing 2,4-dihaloquinazolines
US4138561A (en) 1977-09-30 1979-02-06 Bristol-Myers Company Cyanocarboxamidines and quinazoline process
US4146718A (en) 1978-04-10 1979-03-27 Bristol-Myers Company Alkyl 5,6-dichloro-3,4-dihydro-2(1h)-iminoquinazoline-3-acetate hydrohalides
US4209623A (en) 1978-06-07 1980-06-24 Bristol-Myers Company Pyrimidine-5-N-(1H-tetrazol-5-yl)-carboxamides
US4208521A (en) 1978-07-31 1980-06-17 Bristol-Myers Company Process for the preparation of imidazo[2,1-b]quinazolinones
US4161595A (en) 1978-10-02 1979-07-17 Bristol-Myers Company Levulinic acid salt
DE2845766A1 (en) 1978-10-18 1980-04-30 Schering Ag PYRIDO ANGLE CLAMP ON 2,1-B ANGLE CLAMP ON -CHINAZOLINONE DERIVATIVES, THEIR PRODUCTION AND USE
JPS5653659A (en) 1979-10-09 1981-05-13 Mitsubishi Yuka Yakuhin Kk Blood platelet coagulation suppressing agent
GB2063249A (en) 1979-10-09 1981-06-03 Mitsubishi Yuka Pharma 4-Phenylphthalazine derivatives
US4423075A (en) 1980-06-19 1983-12-27 Ayerst, Mckenna & Harrison Inc. Aldose reductase inhibition by 5-fluoro-2-methyl-1-[[4-(methylsulfonyl)phenyl]methylene]-1H-indene-3-acetic acid
JPS57167974A (en) 1981-04-09 1982-10-16 Mitsubishi Yuka Yakuhin Kk Preparation of 4-phenylphthalazine derivative
DE3131365A1 (en) 1981-08-07 1983-02-24 Henkel KGaA, 4000 Düsseldorf NEW DIGLYCIDYL-SUBSTITUTED HETEROCYCLIC COMPOUNDS, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE IN PHARMACEUTICAL PREPARATIONS WITH CYTOSTATIC EFFECTIVENESS
US5250535A (en) 1982-02-01 1993-10-05 Syntex Inc. Substituted 9-(1 or 3-monoacyloxy or 1,3-diacyloxy-2-propoxymethyl) purines as antiviral agent
US4460591A (en) 1982-08-26 1984-07-17 Sri International 8,10-Dideazaminopterins
US4837239A (en) 1985-08-23 1989-06-06 Syntex (U.S.A.) Inc. Cardiotonic phosphodiesterase inhibitors complexed with water soluble vitamins
ES2031513T3 (en) 1986-08-21 1992-12-16 Pfizer Inc. QUINAZOLINDIONAS AND PIRIDOPIRIMIDINADIONAS.
CA1303037C (en) 1987-02-02 1992-06-09 Smith Kline & French Laboratories Limited Purinone derivatives as bronchodilators vasodilators and anti-allergic agents
EP0330004A1 (en) 1988-02-08 1989-08-30 Schering Corporation Nucleoside-type compounds as phosphodiesterase inhibitors
US5091431A (en) 1988-02-08 1992-02-25 Schering Corporation Phosphodiesterase inhibitors
US5254571A (en) 1988-04-21 1993-10-19 Smith Kline & French Laboratories Ltd. Chemical compounds
GB8814352D0 (en) 1988-06-16 1988-07-20 Smith Kline French Lab Chemical compounds
ES2058527T3 (en) 1988-06-16 1994-11-01 Smith Kline French Lab CONDENSED DERIVATIVES OF PIRIMIDINE PROCEDURE AND INTERMEDIATE COMPOUNDS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.
US5075310A (en) 1988-07-01 1991-12-24 Smith Kline & French Laboratories, Ltd. Pyrimidone derivatives as bronchodilators
US4923874A (en) 1988-07-21 1990-05-08 G. D. Searle & Co. Use of 8-azapurin-6-one derivatives for control of hypertension
GB8817651D0 (en) 1988-07-25 1988-09-01 Smith Kline French Lab Chemical compounds
GB8827988D0 (en) 1988-11-30 1989-01-05 Smith Kline French Lab Chemical compounds
US4971972A (en) 1989-03-23 1990-11-20 Schering Corporation Phosphodiesterase inhibitors having an optionally substituted purine derivative portion and a benzo- or cyclopenta-furan portion
GB8909560D0 (en) 1989-04-26 1989-06-14 Smith Kline French Lab Chemical compounds
US5376683A (en) 1989-07-14 1994-12-27 Schering Aktiengesellschaft Δ8- and Δ9-prostaglandin derivatives, process for their production and their pharmaceutical use
GB8923131D0 (en) 1989-10-13 1989-11-29 Smith Kline French Lab Chemical compounds
US5091430A (en) 1990-03-13 1992-02-25 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services O6 -substituted guanine compounds and methods for depleting O6 -alkylguanine-DNA alkyltransferase levels
GB9013750D0 (en) 1990-06-20 1990-08-08 Pfizer Ltd Therapeutic agents
NZ238609A (en) 1990-06-21 1993-12-23 Schering Corp Polycyclic guanine derivatives; preparation, pharmaceutical compositions,
ZA916646B (en) 1990-08-23 1992-07-29 Synphar Lab Inc Novel isocarbostiryl compounds and anti-tumor use thereof
US5175151A (en) 1990-09-07 1992-12-29 Schering Corporation Antiviral compounds and antihypertensive compounds
AU650689B2 (en) 1990-11-06 1994-06-30 Fgn, Inc. Esters and amides of substituted indenyl acetic acids
AU659107B2 (en) 1990-11-06 1995-05-11 Fgn, Inc. Method for treating colonic polyps by the use of esters and amides of substituted fused ring phenyl acetic acids
AU650914B2 (en) 1990-11-06 1994-07-07 Fgn, Inc. Esters and amides of substituted phenyl acetic acids
AU658373B2 (en) 1990-11-06 1995-04-13 Fgn, Inc. Esters and amides of substituted pyrrole acetic acids
AU659106B2 (en) 1990-11-06 1995-05-11 Fgn, Inc. Method for treating colonic polyps by the use of esters and amides of substituted phenyl and pyridyl amino carboxylates
US5401774A (en) * 1991-03-08 1995-03-28 University Of Arizona Method for treating patients with precancerous lesions by administering substituted sulfonyl idenyl acetic and propionic acids and esters to patients with lesions sensitive to such compounds
AU650720B2 (en) * 1991-03-08 1994-06-30 Fgn, Inc. Method for treating patients with precancerous lesions by administering substituted sulfonyl indenyl acetic and propionic acids and esters thereof
CA2062587A1 (en) 1991-03-11 1992-09-12 Toshiaki Kumazawa Indole derivatives
US5223501A (en) 1991-05-10 1993-06-29 Merck & Co., Inc. Substituted pyrimidinones bearing acidic functional groups as angiotensin ii antagonists
GB9114760D0 (en) 1991-07-09 1991-08-28 Pfizer Ltd Therapeutic agents
PT100905A (en) 1991-09-30 1994-02-28 Eisai Co Ltd BICYCLE HYGIENEOUS HETEROCYCLIC COMPOUNDS CONTAINING BENZENE, CYCLOHEXAN OR PYRIDINE AND PYRIMIDINE, PYRIDINE OR IMIDAZOLE SUBSTITUTES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
GB9121028D0 (en) 1991-10-03 1991-11-13 Pfizer Ltd Therapeutic agents
GB9126260D0 (en) 1991-12-11 1992-02-12 Pfizer Ltd Therapeutic agents
US5731167A (en) 1992-01-17 1998-03-24 The United States Of America As Represented By The Department Of Health And Human Services Autotaxin: motility stimulating protein useful in cancer diagnosis and therapy
JP2657760B2 (en) 1992-07-15 1997-09-24 小野薬品工業株式会社 4-aminoquinazoline derivatives and pharmaceuticals containing them
EP0654024A1 (en) 1992-08-06 1995-05-24 Warner-Lambert Company 2-thioindoles (selenoindoles) and related disulfides (selenides) which inhibit protein tyrosine kinases and which have antitumor properties
DE4330177A1 (en) 1993-08-31 1995-03-02 Schering Ag New 9-chloro prostaglandin derivatives
CA2148260A1 (en) 1993-09-10 1995-03-16 Yasutaka Takase Quinazoline cpompounds
WO1995018969A1 (en) * 1994-01-06 1995-07-13 Terrapin Technologies, Inc. Surrogates for targets and improved reference panels
CA2187200A1 (en) * 1994-04-05 1995-10-12 D. James Morre Nadh oxidase as a target in diagnosis and therapy
US5776962A (en) * 1994-08-03 1998-07-07 Cell Pathways, Inc. Lactone compounds for treating patient with precancerous lesions
US5696159A (en) 1994-08-03 1997-12-09 Cell Pathways, Inc. Lactone compounds for treating patients with precancerous lesions
DE19501481A1 (en) 1995-01-19 1996-07-25 Bayer Ag 2,8-disubstituted quinazolinones
US5674876A (en) 1995-01-20 1997-10-07 Research Development Foundation ρ-heteroatom-substituted phenols and uses thereof
US5614530A (en) 1995-03-10 1997-03-25 Sterling Winthrop Inc. Substituted N-arylmethyl and heterocyclmethyl-1H-pyrazolo[3,4-b]quinolin-4-amines and compositions and methods of use thereof
US5488055A (en) 1995-03-10 1996-01-30 Sanofi Winthrop Inc. Substituted N-cycloalkylmethyl-1H-pyrazolo(3,4-b)quinolin-4 amines and compositions and methods of use thereof
DE19518082A1 (en) 1995-05-17 1996-11-21 Merck Patent Gmbh 4 (-Arylaminomethylene) -2,4-dihydropyrazol-3-one
US5874440A (en) 1995-06-07 1999-02-23 Cell Pathways, Inc. Method of treating a patient having precancerous lesions with phenyl pyrimidinone derivatives
GB9526245D0 (en) 1995-12-21 1996-02-21 Celltech Therapeutics Ltd Chemical compounds
GB9526243D0 (en) 1995-12-21 1996-02-21 Celltech Therapeutics Ltd Chemical compounds
GB9526246D0 (en) 1995-12-21 1996-02-21 Celltech Therapeutics Ltd Chemical compounds
US5798246A (en) 1996-03-25 1998-08-25 Incyte Pharmaceuticals, Inc. Cyclic nucleotide phosphodiesterase
US6063818A (en) * 1996-06-13 2000-05-16 Cell Pathways Inc. Substituted benzylidene indenyl formamides, acetamides and propionamides
US5998477A (en) * 1996-06-13 1999-12-07 Cell Pathways Inc. Substituted methoxy benzylidene indenyl-acetic and propionic acids for treating patients with precancerous lesions
US5869519A (en) 1996-12-16 1999-02-09 Idun Pharmaceuticals, Inc. C-terminal modified (n-substituted)-2-indolyl dipeptides as inhibitors of the ICE/ced-3 family of cysteine proteases
DK0957903T3 (en) 1996-09-18 2005-11-21 Applied Genetics Inc Dermatics Norbornene and norbornanediol compounds for the treatment of pigmentation disorders, neurodegenerative diseases or proliferative skin disorders
US5932465A (en) 1997-10-16 1999-08-03 Icos Corporation Phosphodiesterase 8A
US5922595A (en) 1997-12-09 1999-07-13 Incyte Pharmaceuticals, Inc. Cyclic GMP phosphodiesterase
US5852035A (en) 1997-12-12 1998-12-22 Cell Pathways, Inc. Method for inhibiting neoplastic cells and related conditions by exposure to substituted N- arylmethyl and heterocyclmethyl-1H-pyrazolo (3,4-B) quinolin-4-amines
US5942520A (en) 1998-01-27 1999-08-24 Cell Pathways, Inc. Method for inhibiting neoplastic cells by exposure to substituted N-cycloalkylmethyl-1-H-pyrazolo (3,4-B) quinolone-4 amines

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4115538A (en) * 1976-02-12 1978-09-19 Yamasu Shoyu Kabushiki Kaisha Assay kit for cAMP and/or cGMP and method of using the same
US5760042A (en) * 1985-04-19 1998-06-02 Sankyo Company, Limited Griseolic acid derivatives, their preparation and their use
US5728563A (en) * 1993-01-29 1998-03-17 Taisho Pharmaceutical Co., Ltd. Cyclic nucleotide phosphodiesterase and a method for production thereof
US6156312A (en) * 1993-06-23 2000-12-05 Leskovar; Peter Agents, affecting the hyperactivated immunological effector cells
US6187756B1 (en) * 1996-09-05 2001-02-13 The Massachusetts Institute Of Technology Composition and methods for treatment of neurological disorders and neurodegenerative diseases
US5858694A (en) * 1997-05-30 1999-01-12 Cell Pathways, Inc. Method for identifying compounds for inhibition of cancerous lesions
US6156528A (en) * 1997-05-30 2000-12-05 Cell Pathways, Inc Methods for using a phosphodiesterase in pharmaceutical screening to identify compounds for treatment of neoplasia
US6200771B1 (en) * 1998-10-15 2001-03-13 Cell Pathways, Inc. Method of using a novel phosphodiesterase in pharmaceutical screeing to identify compounds for treatment of neoplasia
US6235782B1 (en) * 1998-11-12 2001-05-22 Rifat Pamukcu Method for treating a patient with neoplasia by treatment with a platinum coordination complex
US6235776B1 (en) * 1998-11-12 2001-05-22 Cell Pathways, Inc. Method for treating a patient with neoplasia by treatment with a paclitaxel derivative

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8271303B2 (en) 2003-09-04 2012-09-18 Hartford Fire Insurance Company System for reducing the risk associated with an insured building structure through the incorporation of selected technologies
US8676612B2 (en) 2003-09-04 2014-03-18 Hartford Fire Insurance Company System for adjusting insurance for a building structure through the incorporation of selected technologies
US10354328B2 (en) 2003-09-04 2019-07-16 Hartford Fire Insurance Company System for processing remote sensor data
US11182861B2 (en) 2003-09-04 2021-11-23 Hartford Fire Insurance Company Structure condition sensor and remediation system

Also Published As

Publication number Publication date
HK1012196A1 (en) 1999-07-30
TW591111B (en) 2004-06-11
TR199800960A3 (en) 1998-12-21
KR19980087436A (en) 1998-12-05
JP3234818B2 (en) 2001-12-04
JP2000198746A (en) 2000-07-18
EP0881300A2 (en) 1998-12-02
KR100304859B1 (en) 2001-11-30
JP3053381B2 (en) 2000-06-19
DE69800488D1 (en) 2001-02-22
CZ295868B6 (en) 2005-11-16
CA2238283A1 (en) 1998-11-30
NO982477D0 (en) 1998-05-29
EP0881300A3 (en) 1999-01-20
EP1038523A2 (en) 2000-09-27
EP1038523A3 (en) 2000-10-25
TR199800960A2 (en) 1998-12-21
DE69800488T2 (en) 2001-08-09
IL124699A0 (en) 1998-12-06
ATE198771T1 (en) 2001-02-15
IL124699A (en) 2003-02-12
AU6979498A (en) 1998-12-10
DE881300T1 (en) 1999-08-19
JPH1194823A (en) 1999-04-09
US6500610B1 (en) 2002-12-31
NO982477L (en) 1998-12-01
ES2132055T1 (en) 1999-08-16
US20030190686A1 (en) 2003-10-09
DK0881300T3 (en) 2001-06-18
EP0881300B1 (en) 2001-01-17
CA2238283C (en) 2002-08-20
JP2000028601A (en) 2000-01-28
AU709666B2 (en) 1999-09-02
NO321717B1 (en) 2006-06-26
CZ165198A3 (en) 1999-01-13
ES2132055T3 (en) 2001-05-01

Similar Documents

Publication Publication Date Title
US6156528A (en) Methods for using a phosphodiesterase in pharmaceutical screening to identify compounds for treatment of neoplasia
EP0881300B1 (en) Method for identifying compounds for inhibition of neoplastic lesions
US6200771B1 (en) Method of using a novel phosphodiesterase in pharmaceutical screeing to identify compounds for treatment of neoplasia
US6130053A (en) Method for selecting compounds for inhibition of neoplastic lesions
US6365627B2 (en) Method for treating a patient with neoplasia by treatment with a paclitaxel derivative
EP0997145B1 (en) Methods for identifying compounds for inhibition of neoplastic lesions
US20030064421A1 (en) Methods for treating a patient with neoplasia by administering cGMP-specific PDE-inhibiting compounds
KR100413591B1 (en) A method for screening compounds for their potential to inhibit neoplasia and pharmaceutical compositions containging such compounds
AU5953899A (en) Method for identifying compounds for inhibition of neoplastic lesions
AU4064402A (en) Method for identifying compounds for inhibition of neoplastic lesions
US6472420B2 (en) Method for treating a patient with neoplasia by treatment with a paclitaxel derivative
KR100644365B1 (en) Methods for identifying compounds for inhibition of neoplastic lesions, and pharmacetical compositions containing such compounds
US20040009464A1 (en) Methods for identifying compounds for inhibition of neoplastic lesions, and pharmacetical compositions containing such compounds
US20030130210A1 (en) Method for treating a patient with neoplasia by treatment with an anthracycline antibiotic
WO2000027193A9 (en) Method for treating a patient with neoplasia by treatment with a gonadotropin releasing hormone analog
WO2002041914A1 (en) Method for treating neoplasia by administering an anti-her2 antibody and a cgmp-specific phosphodiesterase inhibitor

Legal Events

Date Code Title Description
AS Assignment

Owner name: OSI PHARMACEUTICALS, INC., NEW YORK

Free format text: MERGER;ASSIGNOR:CELL PATHWAYS, INC.;REEL/FRAME:014479/0198

Effective date: 20030714

AS Assignment

Owner name: OSI PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CELL PATHWAYS, INC.;REEL/FRAME:014842/0010

Effective date: 20030714

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION