Formulations and methods of using nitric oxide mimetics against a malignant cell phenotype

Abstract
The present invention relates to methods and formulations for inhibiting, treating and preventing a malignant cell phenotype, cell, tumor and/or disease. Administration of nitric oxide mimetics, such as low doses, is sufficient to increase, restore or maintain nitric oxide-mediated signaling in cells so that malignant cell phenotypes, cells, tumors and/or diseases are inhibited or prevented. These methods and formulations are particularly useful in treating and preventing cancer in animals.
Description


FIELD OF THE INVENTION

[0002] The present invention relates to methods and formulations for inhibiting, treating and preventing a malignant cell phenotype, cell, tumor and/or disease. We have now found that the mechanism by which hypoxia and hyponitroxia have impact upon cellular phenotype is not necessarily mediated solely by the lack of oxygen but rather also from a deficiency in nitric oxide mediated signaling. Accordingly, as demonstrated herein, administration of nitric oxide mimetics, such as low doses, is sufficient to increase, restore or maintain nitric oxide-mediated signaling in cells so that malignant cell phenotypes, cells, tumors and/or diseases are inhibited or prevented. Thus, provided herein are formulations and methods of using these formulations to deliver low doses of nitric oxide mimetics to cells at levels which inhibit a malignant cell phenotype, cell, tumor and/or disease to treat and/or prevent development of a malignant cell phenotype, cell, tumor and/or disease but which reduce or avoid development of unwanted effects of the NO mimetics. These methods and formulations are particularly useful in treating and preventing cancer in animals.



BACKGROUND OF THE INVENTION

[0003] Hypoxia or oxygen tension below normal physiologic levels in cells results in physiologic as well as pathologic alterations in the cells, which alterations have been associated with differential gene expression. For example, hypoxia affects endothelial cellular physiology in vivo and in vitro in various ways including modulating the transcriptionally-regulated expression of vasoactive substances and matrix proteins involved in modulating vascular tone or remodeling the vasculature and surrounding tissue (Faller, D. V. Clin. Exp. Pharmacol. and Physiol. 1999 26:74-84). Hypoxia in solid tumors has been shown to protect cancer cells from being killed by X-irradiation and leads to resistance to certain cancer drugs. Hypoxia also appears to accelerate malignant progression and increase metastasis (Brown, J. M. Cancer Res. 1999 59:5863-5870).


[0004] Low oxygen levels have been shown to enhance invasiveness in a number of cell types, and likely promote invasive phenotypes in biological environments such as the core of rapidly growing tumors. Studies in our laboratory suggest that this effect is mediated via a reduction in nitric oxide (NO) production. For example, the inhibition of endogenous NO production (that occurs under hypoxic conditions or in the presence of a NOS inhibitor) was shown to up-regulate both the expression of the urokinase receptor (uPAR, a pro-invasive molecule) and metastasis. Further, the hypoxic upregulation of (uPAR) and metastasis was completely abrogated by the addition of very low concentrations of NO-mimetics.


[0005] Nitric oxide has been implicated in various biological processes. For example, nitric oxide is a biological messenger molecule responsible for endothelium derived vascular relaxation and neurotransmission. Nitric oxide, at what these researchers refer to as high levels, is also known as a mediator for anti-tumor and anti-bacterial actions of macrophages. Nitric oxide has also been demonstrated to play a modulatory role on cytokine-induced expression of matrix metalloproteinase-9 and tissue inhibitors of metalloproteinases (Eberhardt et al. Kidney International 2000 57:59-69).


[0006] Soluble guanylyl cyclase (sGC) is the best defined downstream target of NO. However, since NO is a pleiotropic molecule, it is likely that there are other molecular targets that mediate NO action. For example, it is very likely that NO could attenuate gene expression via currently unknown mechanisms and, as such, impact various cellular functions, such as growth, proliferation, migration, apoptosis, and the like. The heterodimeric protein, i.e., sGC, catalyzes the conversion of guanosine 5′-triphosphate (GTP) to cyclic guanosine monophosphate (cGMP). Nitric oxide binds to the heme moiety of sGC, inducing a number of conformational changes that result in its activation cGMP is a potent second messenger that allows NO to transmit its signal to downstream effectors. Elevated levels of cGMP have been negatively correlated with vascular smooth muscle growth and have been shown to prevent platelet aggregation as well as the adherence of neutrophils to endothelial cells. Further, cGMP levels play a role in gene regulation. A cGMP analogue (8-Br-cGMP) has been shown to prevent the hypoxic induction of VEGF. The endogenous inhibition of endothelin-1 (ET-1) by NO is also mediated through a guanylate cyclase/cGMP-dependent mechanism.


[0007] There are several cGMP target proteins that could mediate NO-regulated gene expression. These include cGMP-dependent protein kinase (PKG), cGMP-activated phosphodiesterases (PDEs), and cGMP-gated ion channels, and under certain conditions, cAMP-dependent protein kinases. Of these, it is thought that PKG is responsible for the majority of cGMP's intracellular actions. PKG is a scrine/threonine protein kinase that is selectively activated by cGMP binding. Upon activation, PKG phosphorylates many intracellular targets, often resulting in alterations in gene expression. For example, PKG has been shown to modulate the transcription of various genes through its activation of factors such as AP-1 and the serum response element. Similarly, Idriss et al. has shown that the c-fos promoter is activated by a PKG dependent mechanism.


[0008] Clinical and experimental data suggest that nitric oxide plays a role in promoting solid tumor growth and progression. For example, nitric oxide generation by inducible nitric oxide synthase (iNOS) has been implicated in the development of prostate cancer (Klotz et al. Cancer; National Library of Medicine, MDX Health Digest 1998 82(10):1897-903), as well as in colonic adenocarcinomas and mammary adenocarcinomas (Lala, P. K. and Orucevic, A., Cancer and Metastasis Reviews 1998 17:91-106). In addition, nitric oxide has been suggested to play an important role in the metabolism and behavior of lung cancers, and in particular adenocarcinomas (Fujimoto et al. Jpn. J. Cancer Res 1997 88: 1190-1198). In fact, it has been suggested that tumor cells producing or exposed to what these researchers refer to as low levels of nitric oxide, or tumor cells capable of resisting nitric oxide-mediated injury undergo a clonal selection because of their survival advantage (Lala, P. K. and Orucevic, A. Cancer and Metastasis Review 1998 17:91-106). These authors suggest that these tumor cells utilize certain nitric oxide-mediated mechanisms for promotion of growth, invasion and metastasis and propose that nitric oxide-blocking drugs may be useful in treating certain human cancers. There is also evidence indicating that tumor-derived nitric oxide promotes tumor angiogenesis as well as invasiveness of certain tumors in animals, including humans (Lala, P. K. Cancer and Metastasis Reviews 1998 17:1-6).


[0009] However, nitric oxide has been reported to reverse production of vasoconstrictors induced by hypoxia (Faller, D. G. Clinical and Experimental Pharmacology and Physiology 1999 26:74-84). In addition, the nitric oxide donors sodium nitroprusside, S-nitroso-L-glutathione and 3-morpholinosydnonimine in the micromolar range (IC50=7.8, 211 and 490 μM, respectively) have been demonstrated to suppress the adaptive cellular response controlled by the transcription factor hypoxia-inducible factor-1 in hypoxically cultured Hep3B cells, a human hepatoma cell line (Sogawa et al. Proc. Natl Acad. Sci. USA 1998 95:7368-7373). The nitric oxide donor sodium nitroprusside (SNP; 150 μM) has also been demonstrated to decrease hypoxia-induced expression of vascular endothelial growth factor, an endothelial cell mitogen required for normal vascular development and pathological angiogenic diseases such as cancer and iris and retinal neovascularization (Ghiso et al. Investigative Ophthalmology & Visual Science 1999 40(6):1033-1039). In these experiments, 150 μM SNP was demonstrated to completely suppress hypoxia-induced VEGF mRNA levels for at least 24 hours in immortalized human retinal epithelial cells.


[0010] High levels of nitric oxide, when induced in certain cells, can cause cytostasis and apoptosis. For example, Xie et al. have demonstrated exposure to high levels of nitric oxide (producing approximately 75 μM nitrite; see FIG. 5A of Xie et al.) to be an exploitable phenomenon to promote death (see FIGS. 6A and 6B of Xie et al.) in murine K-1735 melanoma cells (J. Exp. Med. 1995 181:1333-1343). In addition, WO 93/20806 discloses a method of inducing cell cytostasis or cytotoxicity by exposing cells to a compound such as spermine-bis(nitric oxide) adduct monohydrate at 500 μM which is capable of releasing nitric oxide in an aqueous solution. The compounds are taught to be useful in the treatment of tumor cells as well as in antiparasitic, antifungal and antibacterial treatments. Use of a mega-dosing regimen is suggested, wherein a large dose of the nitric oxide releasing compound is administered, time is allowed for the active compound to act, and then a suitable reagent such as a nitric oxide scavenger is administered to the individual to render the active compound inactive and to stop non-specific damage. It is taught at page 14, line 25-30 of WO 93/20806 that 3-(n-propyl amino)propylamine bis(nitric oxide) adduct, diethylamine-bis(nitric oxide) adduct sodium salt, isopropylamine-bis(nitric oxide) adduct sodium salt, sodium trioxodinitrate (II) monohydrate, and N-nitrosohydroxylamine-N-sulfonate did not significantly affect cell viability at concentrations up to 500 μM.


[0011] U.S. Pat. No. 5,840,759, U.S. Pat. No. 5,837,736, and U.S. Pat. No. 5,814,667, disclose methods for using mg/kg quantities of nitric oxide releasing compounds to sensitize hypoxic cells in a tumor to radiation. These patents also disclose methods of using the same nitric oxide-releasing compounds at mg/kg levels to protect non-cancerous cells or tissue from radiation, to sensitize cancerous cells to chemotherapeutic agents, and to protect non-cancerous cells or tissue from chemotherapeutic agents. Compounds used in these methods spontaneously release nitric oxide under physiologic conditions without requiring oxygen. These patents teach administration of the nitric oxide-releasing compound from about 15 to about 60 minutes prior to therapy. Typical doses of the nitric oxide releasing compound administered are suggested to be from about 0.1 to about 100 mg of one or more nitric oxide releasing compounds per kg of body weight. Concentrations of the nitric oxide releasing compounds DEA/NO and PAPA/NO demonstrated to increase the sensitivity of MCF7 breast cancer cells and V79 fibroblasts to melphalan, thiotepa, mitomycin C, SR4233 and cisplatin in vitro were in the millimolar range while 70 mg/kg of DEA/NO was demonstrated to increase the survival of mice administered the chemotherapeutic agent melphalan in the in vivo KHT tumor model.


[0012] U.S. Pat. No. 5,700,830 and WO 96/15781 disclose methods for inhibiting adherence between cancerous cells and non-cancerous cells in an animal by administering to the animal a nitric oxide-releasing compound containing a nitric oxide-releasing N2O2 functional group. However, studies indicate that cancer cell adhesion to and spreading along the vessel wall leading to extravasation is not an obligatory event in metastasis (Morris et al. Exp. Cell. Res. 1995 219:571-578).


[0013] WO 98/58633 discloses a microdose nitric oxide therapy for alleviating vascular conditions associated with a reduction in nitric oxide production or an attenuation of nitric oxide effect.



SUMMARY OF THE INVENTION

[0014] The present invention provides methods and formulations for administering nitric oxide mimetics to cells to inhibit, to treat and to prevent a malignant cell phenotype, cell, tumor and/or disease. In certain preferred aspects, the nitric oxide mimetic is administered in a low dose. Advantageously, in certain aspects, the nitric oxide mimetics of the present invention increase the efficacy of palliative treatment, by relieving or reducing the intensity of uncomfortable symptoms associated with cancer, such as pain, nausea, and shortness of breath.


[0015] In one aspect, the present invention provides a method useful for controlling, treating, and/or preventing cancer, malignancies, neoplasm, hyperplasia, hypertrophy, dysplasia and/or tumor angiogenesis, comprising administering a nitric oxide mimetic to an animal to control, treat and/or prevent cancer, malignancies, neoplasm, hyperplasia, hypertrophy, metastasis, dysplasia and/or tumor angiogenesis.


[0016] In one embodiment, the nitric oxide mimetic is administered at a low dose. In another embodiment, the nitric oxide mimetic is administered at a level which delays and/or reduces development of tolerance to the nitric oxide mimetic and/or unwanted side effects, including headache, flushing, syncope, dizziness and hypotension. In another embodiment, the nitric oxide mimetic is administered alone or in combination with an antimalignant therapeutic agent. In yet another embodiment, the nitric oxide mimetic is administered in combination with agents intended for palliative purposes including pain relief, improving physical strength and quality of life.


[0017] In another embodiment, the nitric oxide mimetic (1) inhibits the metastatic potential of a malignant cell phenotype, cell, tumor and/or disease preferably by decreasing the invasiveness, progression, growth and/or metastases of cells, tumors and/or diseases exhibiting a malignant phenotype; inhibiting the survival and/or growth of cells, tumors and/or diseases exhibiting a malignant phenotype; decreasing the progression and/or metastases of cells, tumor volume and/or diseases exhibiting a malignant phenotype; increasing the regression of cells, tumors and/or diseases exhibiting a malignant phenotype; and/or facilitating the killing of cells, tumors and/or diseases exhibiting a malignant phenotype; (2) maintains a malignant cell, tumor and/or disease in a dormant state at its primary and/or secondary site; (3) enhances the efficacy of, increases sensitivity to, and/or prevents or decreases the resistance of a malignant cell, tumor and/or disease to an antimalignant therapeutic modality; or (4) inhibits or prevents cell, tumor and/or disease angiogenesis in animals at high risk of developing cancer and/or exposed to factors known to decrease nitric oxide activity in an animal, optionally wherein said factors include decreased arginine levels, exposure to nitric oxide synthase antagonists, exposure to nitric oxide scavengers, changes in nitric oxide synthase expression, change in cofactors, glucose deprivation, surgical procedures, administration of anaesthetic agents, administration of pharmacologic agents which alter circulation, traumatic injuries, physical trauma, blood loss, decreased blood volume, or hemorrhage, or combinations thereof.


[0018] In another embodiment, the cells exhibiting the malignancies are selected from malignant cells, invasive cells, cells and tissue(s) that facilitate the malignant process, and combinations thereof; optionally wherein the malignant cell phenotype is controlled, treated or prevented by improving response to an antimalignant therapeutic modality.


[0019] In another embodiment, the nitric oxide mimetic (1) inhibits or retards the metastasis of existing tumors in a patient preferably by decreasing the invasiveness, progression, growth and/or metastases of existing tumor(s); inhibiting the survival and/or growth of existing tumor(s); decreasing the progression and/or metastases of existing tumor(s); increasing the regression of existing tumor(s); and/or facilitating the killing of existing tumor(s); (2) maintains a malignant tumor in a dormant state at its primary and/or secondary site; (3) enhances the efficacy of, increases sensitivity to, and/or prevents or decreases the resistance of tumor(s) to an antimalignant therapeutic modality; or (4) inhibits or prevents tumor angiogenesis in animals at high risk of developing cancer and/or exposed to factors known to decrease nitric oxide activity in an animal, optionally wherein said factors include decreased arginine levels, exposure to nitric oxide synthase antagonists, exposure to nitric oxide scavengers, changes in nitric oxide synthase expression, change in cofactors, glucose deprivation, surgical procedures, administration of anaesthetic agents, administration of pharmacologic agents which alter circulation, trauimatic injuries, physical trauma, blood loss, decreased blood volume, or hemorrhage, or combinations thereof.


[0020] In another embodiment, the nitric oxide mimetic (1) inhibits or retards the metastasis of a cancerous disease in a patient preferably by decreasing the invasiveness, progression, growth and/or metastasis of the disease; inhibiting the survival and/or growth of the disease; decreasing the progression and/or metastasis of the disease; increasing the regression of the disease; and/or facilitating the curing and/or treatment of the disease; (2) produces or maintains remission of the disease; (3) enhances the efficacy of, increases sensitivity to, and/or prevents or decreases the resistance to an antimalignant therapeutic modality used to treat the disease; or (4) inhibits or prevents development of the disease in animals at high risk of developing cancer and/or exposed to factors known to decrease nitric oxide activity in an animal, optionally wherein the factors include decreased arginine levels, exposure to nitric oxide synthase antagonists, exposure to nitric oxide scavengers, changes in nitric oxide synthase expression, change in cofactors, glucose deprivation, surgical procedures, administration of anaesthetic agents, administration of pharmacologic agents which alter circulation, traumatic injuries, physical trauma, blood loss, decreased blood volume, or hemorrhage, or combinations thereof.


[0021] In another embodiment, the cells exhibiting the malignancies are selected from malignant cells, invasive cells, cells and tissue(s) that facilitate the malignant process, and combinations thereof, optionally wherein the malignant cell phenotype is controlled, treated or prevented by improving response to an antimalignant therapeutic modality.


[0022] In another embodiment, cancer is diagnosed or monitored by measuring a tumor selective marker present in the animal.


[0023] In another embodiment, the nitric oxide mimetic decreases, maintains, or decelerates increases of the level of the tumor marker.


[0024] In another embodiment, the cancer comprises gastric cancer, gastrointestinal cancer, testicular cancer, prostate cancer, prostatic adenocarcinoma, breast cancer, metastatic melanoma, lung cancer, the cancers set forth in Table 2 or combinations thereof. Optionally, the cancer or other malignancies, neoplasm, hyperplasia, hypertrophy, dysplasia and/or tumor angiogenesis in an animal comprises benign prostatic hyperplasia or molar pregnancy.


[0025] In another embodiment, the nitric oxide mimetic comprises nitric oxide, a nitric oxide donor, a nitric oxide prodrug, a compound that generates or releases nitric oxide through biotransformation, a compound that generates nitric oxide spontaneously or spontaneously releases nitric oxide, or a compound which generates nitric oxide, or combinations thereof.


[0026] In another embodiment, the nitric oxide mimetic is (1) a nitric oxide donor selected from nitroglycerin (GTN), isosorbide 5-mononitrate (ISMN), isosorbide dinitrate (ISDN), pentaerythritol tetranitrate (PETN), erthrityl tetranitrate (ETN), N-hydroxyl-L-arginine (NOHA), N6-(1-iminoethyl)lysine) (L-NIL), L-N5-(1-iminoethyl) omithine (LN-NIO), Nw-methyl-L-arginine (L-NMMA), S-nitrosogluthathione (SNOG), S,S-dinitrosodithiol (SSDD), [N-[2 (nitroxyethyl)]-3-pyridinecarboxamide (nicorandil), sodium nitroprusside (SNP), S-nitroso-N-acetylpenicilamine (SNAP), 3-morpholino-sydnonimine (SIN-1), molsidomine, DEA-NONOate (2 (N,N-diethylamino)-diazenolate-2-oxide), and spermine NONOate (N-[4-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]butyl-1,3-propanediamine; (2) a compound that activates stages of NO pathway, a compound which enables or facilitates NO utilization by a cell, a compound which directly activates guanylyl cyclase, or a phosphodiesterase inhibitor, or combinations thereof; (3) a non-specific phosphodiesterase inhibitor, a dual-selective phosphodiesterase inhibitor, a type I, II, III, IV, V, VI, VII, VIII, IX, X, or XI phosphodiesterase inhibitor, or combinations thereof; or (4) a protein kinase G activator.


[0027] In another embodiment, the antimalignant therapeutic agent includes radiation therapy (radiotherapy), thermal therapy, immunotherapy, hormonal therapy, or single agent chemotherapy, combination chemotherapy, chemo-irradiation, adjuvant therapy, neoadjuvant therapy, palliative therapy or combinations thereof. Optionally, the antimalignant therapeutic modality comprises radiation therapy, and the nitric oxide mimetic is a nitric oxide, a nitric oxide donor, a compound that generates or releases nitric oxide through biotransformation, or a compound that generates nitric oxide spontaneously or spontaneously releases nitric oxide only in the presence of oxygen, or combinations thereof, wherein the nitric oxide mimetic is administered during the radiation therapy.


[0028] In another embodiment, the chemotherapy comprises administration of chemotherapeutic agent that is an anti-angiogenic agent, an antimetabolite, an antibiotic, an endothelin activating agent, an enzyme inhibitor, a hormonal agent, ocreotide acetate, a microtubule-disruptor agent, a microtubule-stabilizing agent, a vinca alkaloid, an epipodophyllotoxin, a topoisomerase inhibitor; a prenyl-protein transferase inhibitor, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, a platimum coordination complex, a biological response modifier, a growth factor, an immune modulator, or a monoclonal antibody, or a combination thereof.


[0029] In another embodiment, the palliative therapy comprises glucocorticoid steroids, narcotic pain relief agents, anti-depressants, sex hormones, or a combination thereof.


[0030] In another embodiment, the dose of nitric oxide mimetic is at least 3- to 10,000-fold lower, preferably 100-10,000 fold lower than a dose of nitric oxide mimetic that produces vasodilation.


[0031] In another embodiment, the nitric oxide mimetic is an organic nitrate and the mimetic is administered at a dose of at least 3- to 10,000-fold lower, preferably 100-10,000 fold lower than the dose of nitric oxide mimetic known to produce vasodilation.


[0032] In another embodiment, the nitric oxide mimetic is a known vasodilatory compound and the mimetic is administered at a dose of at least 3- to 10,000-fold lower, preferably 100-10,000 fold lower than the dose of nitric oxide mimetic known to produce vasodilation


[0033] In another embodiment, the nitric oxide mimetic is selected from the group consisting of a calcium channel blocker, an an α-adrenergic receptor antagonist, a β-adrenergic receptor agonist, a phosphodiesterase inhibitor, a cAMP-dependent protein kinase activator, a superoxide scavenger, a potassium channel activator, a benzodiazepine, an adrenergic nerve inhibitor, an antidiarrheal agent, a HMG-CoA reductase inhibitor, an adenosine receptor modulator, a adenylyl cyclase activator, an endothelin receptor antagonist, a bisphosphonate, a cGMP-dependent protein kinase activator, a guanylyl cyclase activator and a SOC inhibitor.


[0034] These methods and formulations are particularly useful in controlling cancer by reducing its growth and improving response to therapy. For example, methods and formulations of the present invention can inhibit metastasis, invasiveness and progression of cells, tumors and/or diseases exhibiting a malignant phenotype. In addition, the methods and formulations can induce or maintain dormancy or remission of cells, tumors and/or diseases exhibiting a malignant phenotype at primary as well as secondary sites. Further, these methods and formulations can prevent or decrease development of resistance of cells, tumors and/or disease exhibiting a malignant phenotype to antimalignant therapeutic modalities, as well as increase the efficacy of antimalignant therapeutic modalities.


[0035] The methods and formulations of the present invention are also very useful in preventing a malignant cell phenotype, cell, tumor and/or disease that can develop upon exposure of cells to conditions and/or therapeutic agents which lead to a deficiency in nitric oxide mimetic activity in the cells.


[0036] The methods and formulations of the present invention are also useful in inhibiting development of a more aggressive malignant cell phenotype in cancer cells, tumors, and/or diseases, which can occur upon exposure to factors which induce such development.


[0037] In addition, these methods and formulations are useful in diagnosing and monitoring a malignant cell phenotype, cell, tumor and/or disease in an animal via detection of levels of one or more markers indicative of a malignant phenotype following administration of a low dose of a nitric oxide mimetic. No change, a decrease or deceleration in the increase of the level of one or more of these markers in an animal following administration of a low dose nitric oxide mimetic as compared to the level of the marker in the animal prior to administration of the low dose nitric oxide mimetic is indicative of a malignant phenotype in the animal. Accordingly, the methods and formulations of the present invention provide new therapeutic and diagnostic approaches for the treatment and prevention of cancer in animals.


[0038] Advantageously, the compounds, and methods described herein prolong cancer remission, prevent the recurrence of cancer, decrease cancer markers, reduce tumor volume, reduce pain, discomfort, and disability (morbidity), increase the quality of life associated with antimalignant therapeutic modalities, decrease cachexia, and reduce the need for anti-emetic agents and narcotic pain killers.







BRIEF DESCRIPTION OF THE DRAWINGS

[0039]
FIG. 1 is a histogram showing the effect of GTN and SNP on the in vitro invasion by MDA-MB-231 invasive breast cancer cells in hypoxic (1% O2) conditions as compared to normal (20% O2) conditions. Cells were coated onto Matrigel-coated membranes and incubated under hypoxic or normal conditions, alone or in the presence of nitric oxide mimetics. The invasion index (% of control), which is taken to be a measure of the invasive potential of the cells for each treatment, was determined by staining the cells that invaded through the membrane and counting them. The first bar depicts the invasion index of cells cultured under normal conditions (20% O2). The second bar depicts the invasion index of cells cultured under hypoxic conditions (1% O2). The third bar depicts the invasion index of cells cultured under hypoxic conditions (1% O2) and administered 10−10 M SNP. The fourth bar depicts the invasion index of cells cultured under hypoxic conditions (1% O2) and administered 10−11 M GTN. The values indicated by “*” were significantly different (p<0.05, n=6) using the Student-Newman-Keuls post-hoc test for pair-wise multiple comparison procedures.


[0040]
FIG. 2 is a histogram showing the lung colonization ability of B16F10 mouse melanoma cells incubated for 12 hours in 1% or 20% O2 in the presence or absence of 2×1031 11 M (20 pM) GTN and injected i.v. (tail vein) into C57B16 female mice. Fourteen days later, mice were sacrificed and lungs were removed and fixed in Bouin's fixative. Both melanotic and amelanotic metastatic colonies were counted under a dissecting microscope. The first bar depicts the number of nodules observed in lungs of mice injected with cells cultured in normal conditions (20% O2). The second bar depicts the number of nodules observed in lungs of mice injected with cells cultured in normal conditions (20% O2) and administered 2×1031 11 M GTN. The third bar depicts the number of nodules observed in lungs of mice injected with cells cultured in hypoxic conditions (1% O2). The fourth bar depicts the number of nodules observed in lungs of mice injected with cells cultured in hypoxic conditions (1% O2) and administered 2×1011 M GTN.


[0041]
FIG. 3 shows circulating prostate specific antigen (PSA) levels in two patients, Patient A (FIG. 3A) and Patient B (FIG. 3B) who had undergone radical prostectomy. A sharp decline in plasma PSA levels was observed in both patients within two months of administration of low dose NO mimetic therapy. In Patient A, this decline continued throughout the course of the study. In Patient B, further increases in PSA levels were minimal. Plasma PSA levels were measured using a radioimmunoassay that has an accuracy of ±0.1 ng/ml.


[0042]
FIG. 4 illustrates the effect of hypoxia (0.5% O2) on cellular cGMP Levels in MDA-MB-231 breast carcinoma cells. Cells were cultured for 6 hrs in 20 or 0.5% O2 and in the presence or absence of GTN (1 mM). IBMX (500 mM) was included in the culture medium in order to inhibit PDE activity, thereby allowing for a measurable accumulation of cGMP. Culture in hypoxic conditions resulted in a 44% reduction in cGMP levels. This effect was completely prevented by GTN (1 mM). Values are presented as fM cGMP per mg of cellular protein +/− standard deviation. Value indicated by an asterisk (*) is significantly different as determined by a one-way analysis of variance followed by the Tukey test for pairwise multiple comparisons (N=3, P<0.05).


[0043]
FIG. 5 illustrates the effect of GTN and SNP on the hypoxic upregulation of in vitro invasion by MDA-MB-231 cells. NO-mimetic drugs were added to the cells at the beginning of the 24-h assay and the invasion index for each treatment was calculated after counting the cells that penetrated through the membrane. Bars represent the mean, normalized, invasion indices±standard deviations. The value indicated by an asterisk (*) was significantly different from the invasion index of non-treated cells incubated in 20% O2 (N=6). P values are indicated in the Examples section below.


[0044]
FIG. 6 illustrates urokinase plasminogen activator receptor (uPAR) expression in MDA-MB-231 breast cancer cells. Western blot analysis of uPAR protein expression by cells incubated in 1% O2 in the presence of various concentrations of glyceryl trinitrate (GTN) (A; N=3) or in 20% O2 in the absence or presence of the NOS antagonist N-monomethyl-L-arginine (L-NMMA, 0.5 μM) alone or in combination with GTN (0.1 nM) (B; N=6). The effect of GTN (1 pM) on uPAR mRNA levels was also examined by Northern blot analysis of cells cultured under conditions similar as for the Western blots (C and D; N=3 and 6 respectively). In all cases, GTN was added at the beginning of the 24-h incubation period. 18S rRNA was used to assess uniformity of RNA loading in the Northern blots. Bars represent means of relative densities±standard deviations. Asterisks (*) indicate statistically significant differences as compared to control (20% O2) values. All P values are indicated in the Examples section below.


[0045]
FIG. 7 illustrates the effect of hypoxia on sGC activity and expression in MDA-MB-231 breast carcinoma cells. (A) Total cellular cGMP accumulated in MDA-MB-231 breast carcinoma cells cultured for 6-h in 20% O2 or 0.5% O2 in the presence or absence of GTN (1 μM). Cyclic GMP levels were measured and normalized to total protein levels. Values represent the mean of accumulated cGMP±standard deviation (N=3). (B) Western blot analysis of sGC expression in cells incubated for 24 hours in 20% O2 or 0.5% O2 (N=3). Bars represent the mean densities±standard deviations. Asterisks (*) indicate statistically significant differences compared to control (20% O2) density values. P values for each condition are indicated in the Examples section below.


[0046]
FIG. 8 illustrates the effect of sGC disruption on the NO-mediated inhibition of uPAR expression in MDA-MB-231 breast carcinoma cells. (A) Northern blot analysis of uPAR mRNA expression in cells cultured for 24 hours in 20% O2 or 0.5% O2, with or without GTN (1 μM) and the selective sGC inhibitor (ODG, 0.5 μM; N=5). (B) Northern blot analysis of uPAR expression in cells cultured for 24-h in the presence or absence of the heme disrupter DFO (100 mM), alone or in combination with GTN (1 μM) (N=5). Bars in both (A) and (B) represent mean relative densities±standard deviations. Values indicated by an asterisk (*) are significantly different. All P values are indicated in the Examples section below.


[0047]
FIG. 9 illustrates the effect of 8-Br-cGMP on hypoxia- and DFO-induced uPAR expression in MDA-MB-231 breast carcinoma cells. (A) Northern blot analysis of uPAR expression in cells cultured for 24 hours in 20% O2 or 0.5% O2 in the absence or presence of various concentrations of 8-Br-cGMP (N=3). (B) Western blot analysis of uPAR protein in cells cultured for 24-h in 20% O2 or 0.5% O2 in the absence or presence of various concentrations of 8-Br-cGMP (N=9). (C) Northern blot analysis of uPAR transcript in cells cultured for 24-h in the absence or presence of DFO (100 μM) alone or with 8-Br-cGMP (1 μM) (N=6). Bars in (A), (B), and (C) represent mean relative densities±standard deviations. Values indicated by an asterisk (*) are significantly different from the control value (20% O2). All P values are indicated in the Examples section below.


[0048]
FIG. 10 illustrates the effect of PKG inhibition on uPAR expression in MDA-MB-231 breast carcinoma cells. (A) Northern blot analysis of uPAR expression in cells cultured for 6-h in the presence or absence of a PKG inhibitor (KT5823, 10 μM) (N=6). (B) Western blot analysis of uPAR protein levels in cells cultured for 6-h in the presence or absence of a PKG inhibitor (KT5823, 10 μM) (N=6). Bars represent mean relative density±standard deviation. Values indicated by an asterisk (*) are significantly different from the control value (20% O2). All P values are indicated in the Examples section below.


[0049]
FIG. 11 illustrates the effect of 8-Br-cGMP on the hypoxic upregulation of the in vitro invasiveness of MDA-MB-231 breast carcinoma cells. Cells were allowed to invade through a reconstituted extracellular matrix for 24-h in 20% O2 or 0.5% O2 in the absence or presence of varying concentrations of 8-Br-cGMP. The invasion index for each treatment was calculated after counting the cells that penetrated through the membrane. Values are presented as the relative mean of the invasion indices±standard deviation. The value indicated by an asterisk (*) was significantly different from the invasion index of the control (20% O2) cells (N=6). All P values are indicated in the Examples section below.


[0050]
FIG. 12 illustrates the effect doxorubicin (12.5-100 μM) on survival of human PC-3 prostate cancer cells. Cells were pre-exposed to 20% or 0.5% O2 for 24 h before a 1 hr treatment with doxorubicin. Survival was assessed by clonogenic ability.


[0051]
FIG. 13 illustrates the effect of a single dose of GTN on the hypoxia-induced resistance to doxorubicin. Treatment of (A) human PC-3 and (B) TRAMP-C2 cells with GTN (0.1 nM) prevented the acquisition of hypoxia-induced resistance to 12.5 μM doxorubicin over a 12 hour incubation in hypoxia. Results are presented as means±standard deviation (N=12). Single asterisk (*) indicates a statistically significant difference (P<0.0001) in survival compared with cells incubated in 20% O2 at 12.5 μM doxorubicin. Double asterisk (**) indicates a statistically significant difference (P<0.0001) in survival compared with cells incubated in 0.5% O2 alone.


[0052]
FIG. 14 illustrates the effect of MDA-MB-231 human breast carcinoma cell spheroid culture on resistance to doxorubicin over time. Following initial cell plating, spheroids formed for 24, 48, 72, 96 or 120 h. At each time point, spheroids were exposed to 200 μM of doxorubicin, then disaggregated and plated for assessment of colony formation (survival). At this concentration of doxorubicin, survival in monolayer culture was zero (not shown).


[0053]
FIG. 15 illustrates the effect of 200 μM doxorubicin in combination with the NO donor drug DETA/NO on MDA-MB-231 breast carcinoma spheroid cell survival. Spheroids were allowed to form for 72 h. DETA/NO was applied for the final 24 h of spheroid formation. Spheroids were then exposed to doxorubicin for 1 h, prior to disaggregation and plating for assessment of survival by clonogenic assay.


[0054]
FIG. 16 illustrates the effect of GTN therapy on a patient with prostate cancer wherein the prostate is still intact. PSA levels in one patient with prostate cancer wherein the prostate is still intact are shown. This patient was administered three episodes of treatment, for approximately one month each, of GTN, 0.03 mg/hour, 24 hours a day. As shown, following commencement of the first two episodes, a decrease in the rate of increase in PSA levels was observed. Following commencement of the third episode, a decrease in PSA levels was observed.


[0055]
FIG. 17 illustrates the effect of GTN therapy in combination with radiation therapy. Circulating PSA levels in one patient with prostate cancer wherein the prostate is still intact are shown. This patient was administered GTN chronically, transdermally at a concentration of 0.03 mg/hour. Two months after chronic GTN therapy was begun, the patient was administered radiation therapy. As shown, this combination therapy accelerated the rate of PSA decrease to within three months. The expected average for a similar decrease in PSA levels following radiation therapy alone is twelve months.







DETAILED DESCRIPTION OF THE INVENTION

[0056] We have now demonstrated that the mechanism by which hypoxia and hyponitroxia have impact on cellular phenotypes is not mediated solely due to a lack of oxygen, but rather also from a deficiency in nitric oxide mimetic activity. Further, we have now demonstrated that administration of a low dose of a nitric oxide mimetic is sufficient to increase, restore or maintain levels of nitric oxide mimetic activity of cells so that a malignant cell phenotype is inhibited or prevented. This inhibition and prevention occurs even when the cells are in a hypoxic environment and/or when combined with inhibition of endogenous nitric oxide production. Nitric oxide mimetics are effective under normoxic conditions as well as hypoxic conditions. Administration of very low doses of nitric oxide mimetics, even under conditions of markedly reduced levels of oxygen (1% O2), was able to prevent the generation of a malignant cell phenotype and inhibit a malignant cell phenotype of cells.


[0057] Accordingly, the present invention relates to the use of nitric oxide mimetic (e.g., low dose) therapy in inhibiting and preventing a malignant cell phenotype of cells. The methods and formulations of the present invention provide new therapeutic approaches for the treatment and prevention of cancer in animals. For purposes of the present invention, by “treatment” or “treating” it is meant to encompass all means for controlling cancer by reducing growth of cells exhibiting a malignant cell phenotype and improving response to antimalignant therapeutic modalities. Thus, by “treatment” or “treating” it is meant to inhibit the survival and/or growth of cells exhibiting a malignant cell phenotype, prevent the survival and/or growth of cells exhibiting a malignant cell phenotype, decrease the invasiveness of cells exhibiting a malignant cell phenotype, decrease the progression of cells exhibiting a malignant cell phenotype, decrease the metastases of cells exhibiting a malignant cell phenotype, increase the regression of cells exhibiting a malignant cell phenotype, and/or facilitate the killing of cells exhibiting a malignant cell phenotype. “Treatment” or “treating” is also meant to encompass maintenance of cells exhibiting a malignant cell phenotype in a dormant (or quiescent) state at their primary site as well as secondary sites. Further, by “treating or “treatment” it is meant to increase the efficacy as well as prevent or decrease resistance to antimalignant therapeutic modalities. By “antimalignant therapeutic modalities” it is meant to include, but is not limited to, radiation therapies, thermal therapies, immunotherapies, hormone therapies, single agent chemotherapies, combination chemotherapies, chemo-irradiation therapies, adjuvant therapies, neo-adjuvant therapies, palliative therapies, and other therapies used by those of skill in the art in the treatment of cancer and other malignancies. “Treating” or “treatment” is also meant to encompass prolonged cancer remission, prevention of recurrence, decrease of cancer markers, reduction in cancer volume, reduction of pain, discomfort, and disability (morbidity), increase in quality of life associated with antimalignant therapeutic modalities, a decrease in mucositis, and a reduction in the need for anti-emetic agents and narcotic pain killers. By “increasing the efficacy”, it is meant to include an increase in potency and/or activity of the antimalignant therapeutic modality and/or a decrease in the development of resistance to the antimalignant therapeutic modality, and/or an increase in sensitivity of the malignant cells and/or tumor to the antimalignant therapeutic modality.


[0058] The present invention also relates to methods of monitoring and/or diagnosing malignant cell phenotypes in an animal via measurement of tumor selective markers in an animal in the presence of NO mimetic (e.g. low dose) therapy. Exemplary tumor markers useful in the monitoring and diagnosing of tumor progression and metastases include, but are not limited to, prostate specific antigen (PSA) for prostate cancer, carcinoembryonic antigen (CEA) and polypeptides such as gastrin and glucagon for gastrointestinal cancer, α-fetoprotein (AFP) and βhCG for testicular cancer, α-fetoprotein (AFP), human chorionic gonadotrophin (HCG) and lactate hydroginase (LDH) in germ cell cancers, HCG in choriocarcinoma, serum AFP in hepatocellular carcinoma, neuron-specific enolase (NSE) in small-cell lung cancer, paraprotein levels and B2-microglobulin which may be of prognostic value in myeloma, 5-hydroxyindole acetic acid (5HIAA) urine levels in carcinoid tumors, squamous cell carcinoma antigen (SCC) and cytokeratin fragments for squamous cell carcinoma, SCC and CA 125 for prognostic information in cervical carcinoma, CA19-9 and CA72-4 for gastric cancer, CD 24 for various cancers, including p-selectin ligand in non-small cell lung cancer, NFκB for prostate cancer prognosis, serum tumor marker CA125 for ovarian cancer, CDKN2A deletion as a diagnostic marker for malignant mesothelioma, dysadherin overexpression in pancreatic ductal adenocarcinoma, SMAD7 as a prognostic marker in patients with colorectal cancer, TGFβ1 as a potential prognostic marker for breast cancer patients with advanced disease, neuroendocrine and cytokeratin serum markers as prognostic determinants of small cell lung cancer, ErbB2 and bone sialoprotein as markers for metastatic osteosarcoma cells, cyclin E and CA15-3 for breast cancer and the cell surface receptors for estrogen and Her-2 for breast cancer. Additional markers which can be monitored for diagnostic purposes include, but are not limited to, calcitonin and calcitonin-related peptide for diagnosis and screening of medullary thyroid carcinoma, Protein Regulated by OXYgen-1 (PROXY-1), also known as NDRG-1, plasminogen activator inhibitor (PAI-1), urokinase-type plasminogen activator receptor (uPAR) and vascular endothelial growth factor (VEGF). Further, as will be understood by those of skill in the art upon reading this disclosure, additional tumor markers to those exemplified herein can also be monitored in the present invention. In a preferred embodiment, the tumor marker is detectable in a biological fluid such a serum, plasma or urine. No change, a decrease or deceleration in the increase of the level of one or more of these markers in an animal following administration of a low dose nitric oxide mimetic as compared to the level of the marker in the animal prior to administration of the low dose nitric oxide mimetic is indicative of a malignant cell phenotype in the animal.


[0059] For purposes of the present invention by the term “low dose” it is meant an amount of nitric oxide mimetic which is capable of increasing, restoring or maintaining a level of nitric oxide mimetic activity to cells, tumors and/or diseases which inhibits or prevents malignant cell phenotypes and/or which increases efficacy of an antimalignant therapeutic modality co-administered with the low dose NO mimetic. At this low dose, the known untoward effects of NO mimetics in animals without a malignant cell phenotype, cell, tumor and/or disease do not occur. As will be understood by those of skill in the art upon reading this disclosure, the nitric oxide mimetic increases, restores or maintains activity both in and around the cell (i.e. in the cellular microenvironment).


[0060] Methods for determining levels of nitric oxide of cells based upon nitrite, nitrate and S-nitrosothiol levels in cell culture, as well as plasma and serum, have been described. Serum or plasma nitrate levels in healthy normal volunteers have been reported to show a mean nitric oxide level of 33.4±8.9 μM with a range of 14 to 60 μM (Marzinzig et al. Nitric Oxide: Biology and Chemistry 1987 1(2): 177-189). These levels, however, are based on NO synthase end products, which accumulate and thus are likely to represent an overestimate of normal physiologic nitric oxide levels. Reported measured levels also vary depending upon the method selected for measurement. Further, levels of nitrite and nitrate in the plasma or serum are not solely representative of a patient's NO production. Based upon our experiments, we believe that normal physiologic levels of nitric oxide mimetic activity of cells may be lower, for example at least 5-fold, and preferably 10- to 10,000-fold lower, than those reported in the art, depending upon the cell.


[0061] Short-term nitric oxide mimetic therapy is generally administered at levels which increase nitric oxide mimetic activity of cells above normal physiologic levels. For purposes of the present invention, however, wherein longer-term therapy is generally desired, induction of tolerance against the NO mimetic and side effects become concerns. Thus, in certain aspects, the amount of nitric oxide mimetic administered is preferably very low so as to delay and/or reduce development of tolerance to the administered NO mimetic and/or unwanted side effects. For example, it is known that administration of nitric oxide or compounds which deliver nitric oxide to human beings at doses conventionally employed to treat cardiovascular conditions (i.e. GTN at 0.2 mg/h or greater) by vasodilation can provoke powerful vasodilator responses as well as development of drug tolerance against GTN upon repeated administration. Such administration is often accompanied by a number of undesirable side effects including headache, flushing and hypotension. In contrast, preferred doses of nitric oxide mimetic administered in the present invention to inhibit and prevent a malignant cell phenotype, cell, tumor and/or disease are lower, preferably at least 3 to 10,000-fold lower, more preferably at least 100- to at least 10,000-fold lower than those typically used in other therapeutic applications such as vasodilation and thus do not induce tolerance to the NO mimetic as quickly nor undesirable side effects. For example, using the nitric oxide mimetics sodium nitroprusside (SNP) and glyceryl trinitrate (GTN), we have now demonstrated that amounts ranging between 10−12 and 10−10 M in the cellular environment can be used to prevent and inhibit a malignant cell phenotype, cell, tumor and/or disease. Further, based on results from these experiments, we believe that doses of SNP as low as 10−14 M would be effective in preventing and inhibiting a malignant cell phenotype, cell, tumor and/or disease in less hypoxic or hyponitroxic environments. Table 1 provides additional examples of various lower preferred doses for nitric oxide mimetics useful in the present invention as well as the comparative higher doses used in vasodilation therapy.


[0062] As will be understood by those of skill in the art upon reading this disclosure, lower or higher amounts of nitric oxide mimetics than those exemplified herein can also be administered based upon the efficacy of the nitric oxide mimetic in achieving the ultimate goal of increasing, restoring or maintaining nitric oxide mimetic activity of cells so that a malignant phenotype is prevented or inhibited without substantial drug tolerance to the NO mimetic developing and without unwanted side effects. Determining amounts of nitric oxide mimetic to be incorporated into the low dose formulations of the present invention can be performed routinely by those skilled in the art based upon the teachings provided herein.


[0063] By the phrase “inhibiting and preventing” as used herein, it is meant to reduce, reverse or alleviate, ameliorate, normalize, control or manage a biological condition. Thus, inhibiting and preventing a malignant cell phenotype in accordance with the present invention refers to preventing development, reversing or ameliorating development and/or normalizing, controlling or managing development of a malignant cell phenotype, cell, tumor and/or disease. Additionally inhibiting and preventing a malignant tumor in accordance with the present invention refers to preventing development, reversing or ameliorating development and/or normalizing, controlling or managing development of a malignant tumor. Similarly, inhibiting and preventing a malignant disease in accordance with the present invention refers to preventing development, reversing the ameliorating development and/or normalizing, controlling or managing development of a malignant disease. Accordingly, administration of a low dose of a nitric oxide mimetic can be used both (1) prophylactically to inhibit and prevent a malignant cell phenotype, cell, tumor and/or disease from developing in animals at high risk for developing cancer or exposed to a factor known to decrease nitric oxide mimetic activity of cells, and (2) to treat cancer in animals by inhibiting metastases and development of resistance to antimalignant therapeutic modalities and increasing the efficacy of antimalignant therapeutic modalities. According to Stedman's Medical Dictionary, malignant is defined as 1) Resistant to treatment; occurring in severe form, and frequently fatal; tending to become worse and lead to an ingravescent course; and 2) in reference to a neoplasm, having the property of locally invasive and destructive growth and metastasis. In accordance with this definition, for purposes of the present invention, by “malignant cell phenotype” it is meant to encompass increases in metastasis, resistance to antimalignant therapeutic modalities, and angiogenesis. By “malignant cell phenotype, cell, tumor and/or disease” for purposes of the present invention, it is also meant to be inclusive of conditions in the spectrum leading to malignant behavior and abnormal invasiveness such as hyperplasia, hypertrophy and dysplasia, as well as those cells and tissue that facilitate the malignant process. Examples of conditions in this spectrum include, but are not limited to, benign prostatic hyperplasia and molar pregnancy. As evidenced by data presented herein, inhibition and prevention of a malignant cell phenotype in cells, tumors and/or diseases can be routinely determined by examining expression of genes including, but not limited to, uPAR, PSA, PAI-1, PROXY-1 and VEGF, by examining cell invasiveness in in vitro or in vivo assays and/or by examining resistance of the cells to antimalignant therapeutic modalities. It is believed that elevated phosphodiesterase expression and/or activity may be observed in cells with a malignant cell phenotype. Methods for measuring expression of these genes have been described for example in WO 99/57306, which is herein incorporated by reference. As will be understood by those of skill in the art upon reading this disclosure, however, other methods for determining gene expression via measurement of expressed protein or proteolytic fragments thereof can also be used. For purposes of the present invention, by the term “nitric oxide mimetic” it is meant nitric oxide, or a functional equivalent thereof; any compound which mimics the effects of nitric oxide, generates or releases nitric oxide through biotransformation, generates nitric oxide spontaneously, or spontaneously releases nitric oxide; any compound which in any other manner generates nitric oxide or a nitric oxide-like moiety or activates other stages of the NO pathway; or any compound which enables or facilitates NO utilization by the cell, when administered to an animal. Such compounds can also be referred to as “NO donors”, “NO prodrugs”, “NO producing agents”, NO delivering compounds”, NO generating agents”, NO releasing agents, and “NO providers”. Examples of such compounds include, but are not limited to: organonitrates such as nitroglycerin (GTN), isosorbide 5-mononitrate (ISMN), isosorbide dinitrate (ISDN), pentaerythritol tetranitrate (PETN), erthrityl tetranitrate (ETN); amino acid derivatives such as N-hydroxyl-L-arginine (NOHA), N6-(1-iminoethyl)lysine) (L-NIL), L-N5-(1-iminoethyl)ornithine (LN-NIO), Nω-methyl-L-arginine (L-NMMA), and S-nitrosoglutathione (SNOG);



Table 1


Typical Vasodilatory and Low Doses of Organonitrate

[0064]

1





TABLE 1










Typical Vasodilatory and Low Doses of Organonitrate










Compound
Commercial Product
Vasodilatory Dose
Preferred Dose According to the Present Invention





Nitroglycerin
Nitrostat ® (Parke-Davis); 0.3 mg, 0.4
Dissolve one tablet (0.3-0.6 mg)
Dissolve one tablet containing from about 0.02 μg to


(sublingual
mg and 0.6 mg sublingual tablets
sublingually or in the buccal
about 0.1 mg sublingually or in the buccal pouch


tablets)

pouch at the first sign of an acute




anginal attack


Nitroglycerin
Nitrolingual ® Spray (Rhone-Poulenc
One or two metered doses
About 0.02 μg to about 0.1 mg sprayed onto or under


(lingual
Rorer); metered aerosol, 0.4
(0.4-0.8 mg) sprayed onto or under
the tongue


aerosol)
mg/metered dose
the tongue at the onset of an




anginal attack


Nitroglycerin
Minitran ® (3M Corporation);
Suggested dose is between 0.2-0.8
About 0.0125 μg/hr-0.1 mg/h


(transdermal
Transdermal patches having the
mg/h for 12-14 h daily with a


patch)
following characteristics (size (cm2),
minimum nitrate-free interval of



delivery rate (mg/h)); (3.3, 0.1; 6.7, 0.2;
10-12 h



13.3, 0.4; and 20.0, 0.6)


Nitroglycerin
NITRO-BID ® Ointment (Hoechst
Doses used in clinical trials have
Ointment containing about 0.375 μg to about 3.75 mg


(ointment)
Marion Roussel); lactose and 2%
ranged from ½ inch (1.3 cm; 7.5
of nitroglycerin applied to the arms or legs over an



nitroglycerin in a base of lanolin and
mg), to 2 inches (5.1 cm; 30 mg),
area of about 36 square inches (232 cm2)



white petrolatum. Each inch (2.5 cm),
typically applied to 36 square



as squeezed from the tube, contains
inches (232 square cm) of skin on



approximately 15 mg of nitroglycerin
the arms or legs


Isosorbide 5-
IMSO ® (Wyeth-Ayerst) 20 mg tablets
20 mg twice daily
About 1 μg to about 2.5 mg twice daily


mononitrate


Erythrityl
Cardilate ® (Burroughs-Wellcome);
Chronic (Adults): 10 mg orally 4
Chronic (Adults): About 0.5 μg to about 1.25 mg


tetranitrate
oral/sublingual tablets, 5 mg, 10 mg
times daily, gradually increased to
orally 4 times daily, gradually increased to about 1 μg




20 mg, if necessary, not to exceed
to about 2.5 mg/day, if necessary, not to exceed about




100 mg/day.
5 to about 12.5 mg/day


Sodium
Nipride ® (Roche); Nitropress ®
Slow infusion at a rate of 0.5
Slow infusion at a rate of from 0.025 ng/kg/min to


nitroprusside
(Abbott); intravenous solution
μg/kg/mn of a solution of 50 mg
about 0.063 μg/kg/min of a solution of 50 mg in




in 500-1000 mL of 5% dextrose
500-1000 mL of 5% dextrose up to a limit of about 0.18




up to a limit of 3.5 mg/kg in brief
mg/kg to about 0.44 mg/kg in brief infusions




infusions


Molsidomine
Corvaton ® (Hoechst Marion Roussel);
2 mg/day up to 36 mg/day given
0.1 μg/day up to 4.5 mg/day given in separate doses



2 mg, 4 mg, and 6 mg tablets
in separate doses either twice or
either twice or three times daily




three times daily


Nicorandil
Nicorandil ® (Chugai Pharmaceuticals,
For the treatment of angina 10-20
About 0.5 μg to about 1 mg twice daily



Japan), Dancor ® (Merck) 10 mg, 20
mg twice daily



mg tablets










[0065] NO prednisone and other compounds which generate or release NO under physiologic conditions such as S,S-dinitrosodithiol (SSDD), [N-[2-(nitroxyethyl)]-3-pyridinecarboxamide (nicorandil), sodium nitroprusside (SNP), S-nitroso-N-acetylpenicilamine (SNAP), 3-morpholino-sydnonimine (SIN-1), molsidomine, DEA-NONOate(2-(N,N-diethylamino)-diazenolate-2-oxide), and spermine NONOate (N-[4-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]butyl-1,3-propanediamine). Organic nitrates GTN, ISMN, ISDN, ETN, and PETN, as well as nicorandil (commonly known as a potassium channel opener) are commercially available in pharmaceutical dosage forms. SIN-1, SNAP, S-thioglutathione, L-NMMA, L-NIL, L-NIO, spermine NONOate, and DEA-NONOate are commercially available from Biotium, Inc. Richmond, Calif. As used herein the term “nitric oxide mimetic” is also intended to mean any compound which acts as a nitric oxide pathway mimetic, that has nitric oxide-like activity, or that mimics the effect of nitric oxide. Such compounds may not necessarily release, generate or provide nitric oxide, but they have a similar effect to nitric oxide on a pathway that is affected by nitric oxide. For example, nitric oxide has both cyclic GMP-dependent and cyclic GMP-independent effects. Nitric oxide is known to activate the soluble form of guanylyl cyclase thereby increasing intracellular levels of the second messenger cyclic GMP and other interactions with other intracellular second messengers such as cyclic AMP. As such, compounds which directly activate either particulate or soluble guanylyl cyclase such as natriuretic peptides (ANP, BNP, and CNP), 3-(5′-hydroxymethyl-2′furyl)-1-benzyl indazole (YC-cGMP or YC-1) and 8-(4-chlorophenylthio) guanosine 3′,5′-cyclic monophosphate (8-PCPT-cGMP), are also examples of NO-mimetics. In some embodiments of the present invention, however, it is preferred that the NO-mimetic not encompass a compound which directly activates either particulate or soluble guanylyl cyclase. Nitric oxide mimetic activity encompasses those signal transduction processes or pathways which comprise at least one NO mimetic-binding effector molecule, such as for example, guanylyl cyclase and other heme containing proteins. Examples of agents which function as NO mimetics by enabling or facilitating NO utilization by the cell are compounds which inhibit phosphodiesterase activity and/or expression, such as phosphodiesterase inhibitors.


[0066] In a preferred embodiment of the present invention, more than one NO mimetic is administered. In this embodiment, it is preferred that the NO mimetics target or act upon different parts of the NO pathway of the cell. For example, an NO donor can be co-administered with a compound that inhibits cyclic nucleotide (e.g. cAMP or cGMP) degradation such as a phosphodiesterase inhibitor. Preferred phosphodiesterase (PDE) inhibitors useful as NO mimetics are those inhibiting PDE-1 through PDE-11.


[0067] By the term “hyponitroxia” in the present invention, it is meant conditions where levels of nitric oxide mimetic activity are lower than normal physiologic levels for that cell type.


[0068] Certain compounds suitable for use in the present invention are well known in the art and are described, e.g., in Goodman and Gilman, The Pharmacological Basis of Therapeutics (9th Ed.), McGraw-Hill, Inc. (1996); The Merck Index (12th Ed.), Merck & Co., Inc. (1996); The Physician 's Desk Reference (49th Ed.), Medical Economics (1995); and Drug Facts and Comparisons, Facts and Comparisons (1993).


[0069] NO Donors


[0070] In preferred embodiments, the compounds of the present invention are NO donors. The nitric oxide donor can be any of a variety of NO donors including, for example, organic NO donors, inorganic NO donors and prodrug forms of NO donors. Additional suitable NO donors include compounds that can be metabolized in vivo into a compound which delivers nitric oxide (e.g., a prodrug form of a NO donor; a NO-releasing drug such as a NO-releasing non-steroidal anti-inflammatory drug (NO-NSAIDs), examples of which include nitro-aspirin, NCX 4016, nitro-(flurbiprofen), HCT 1026, NCX 2216; or a binary NO generating system, such as acidified nitrates), or compounds that serve as physiological precursor of nitric oxide, such as L-arginine and salts of L-arginine. The NO donor may include at least one organic nitrate (including esters of nitric acid) and can be either a cyclic or acyclic compound. For example, suitable NO donors include nitroglycerin (NTG), isosorbide dinitrate (ISDN), isosorbide mononitrate (ISMN) which may include isosorbide-2-mononitrate (IS2N) and/or isosorbide-5-mononitrate (IS5N), erythrityl tetranitrate (ETN), pentaerythritol tetranitrate (PETN), ethylene glycol dinitrate, isopropyl nitrate, glyceryl-1-mononitrate; glyceryl-1,2-dinitrate, glyceryl-1,3-dinitrate, butane-1,2,4-triol trinitrate, and the like. Nitroglycerin and other organic nitrates including ISDN, ETN, and PETN, have been given regulatory approval for use in treatments in other fields of medicine on human subjects. Additional NO donors include sodium nitroprusside, N,O-diacetyl-N-hydroxy-4-chlorobenzenesulfonamide, NG-hydroxy-L-arginine (NOHA), hydroxyguanidine sulfate, molsidomine, 3-morpholinosydnonimine (SIN-1), (±)-S-nitroso-N-acetylpenicillamine (SNAP), S-nitrosoglutathione (GSNO), (±)-(E)-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexeneamide (FK409), (±)-N-[(E)-4-ethyl-3-[(Z)-hydroxyimino]-5-nitro-3-hexen-1-yl]-3-pyridinecarboxamide (FR144420), and 4-hydroxymethyl-3-furoxancarboxamide. In addition, compounds that interfere with the breakdown of NO in vivo may be administered.


[0071] Additional NO Mimetics


[0072] In certain embodiments, the compounds and methods of the present invention are not limited to the foregoing traditional nitric oxide mimetics. As explained in detail below, these nitric oxide mimetics compounds include, for example, calcium channel blockers, α-adrenergic receptor antagonists and β-adrenergic receptor agonists, phosphodiesterase inhibitors, cAMP-dependent protein kinase activators, superoxide scavengers, potassium channel activators, benzodiazepines, adrenergic nerve inhibitors, antidiarrheal agents, HMG-CoA reductase inhibitors, adenosine receptor modulators, adenylyl cyclase activators, endothelin receptor antagonists, bisphosphonates, cGMP-dependent protein kinase activators, guanylyl cylase activators and SOC inhibitors. In certain aspects, the compounds are not limited to a low dose. Although a low dose can of course be used, the dosing of these compounds is not so limited. The compounds set forth below (e.g., PDE inhibitors) can be used at various doses such as high and low doses.


[0073] Calcium Channel Blockers


[0074] Ca2+ channel blockers are compounds that inhibit the entry of Ca2+ into the cell from the extracellular fluid. Suitable Ca2+ channel blockers for use with the methods of the present invention include, but are not limited to, nifedipine, nimodipine, felopidine, nicardipine, isradipine, amlodipine, diltiazem, bepridil, verapamil etc. (see, e.g., WO 98/36733). L-type Ca2+ channel blockers are also available.


[0075] α-adrenergic Receptor Antagonists and β-adrenergic Receptor Agonists


[0076] Additional preferred compounds for use in the context of the present invention include, e.g., α-adrenergic receptor antagonists and β-adrenergic receptor agonists. Suitable α-adrenergic receptor antagonists include, for example, α1-adrenergic receptor antagonists, α2-adrenergic receptor antagonists and other nonspecific α-adrenergic receptor antagonists. Preferred α1-adrenergic receptor antagonists include, but are not limited to, prazosin, doxazosin, phenoxybenzamine, phentolamine, terazosin, tolazoline, etc., and are described in Goodman and Gilman, “The Pharmaceutical Basis of Therapeutics,” 9th Edition, Hardman, et al. (ed.), McGraw-Hill (1996). Suitable α2-adrenergic receptor antagonists include, but are not limited to, yohimbine and are also described in Goodman and Gilman, “The Pharmaceutical Basis of Therapeutics,” 9th Edition, Hardman, et al. (ed.), McGraw-Hill (1996). Other suitable antagonists are α2-adrenergic antagonists include, for example, post-synaptic α2-adrenergic antagonists. These post-synaptic α2-adrenergic antagonists include, but are not limited to, imiloxan, ARC 239 dihydrochloride and other pharmaceutically acceptable salts thereof. ARC 239 dihydrochloride is 2-[2-(4-(2-Methoxyphenyl)piperazin-1-yl)ethyl]-4,4-dimethyl-1,3-(2H,4H)-isoquinolindone dihydrochloride. Other suitable post-synaptic α2-adrenergic antagonists include, but are not limited to, idazoxan, rauwolscine, efaroxan, mianserin, and mirtazapine. Of these, mianserin and mirtazapine are particularly preferred. Suitable β-adrenergic receptor agonists for use with the methods of the present invention include, but are not limited to, β1-adrenergic receptor agonists, β2-adrenergic receptor agonists, β3-adrenergic receptor agonists and other nonspecific β-adrenergic receptor agonists. In a preferred embodiment, the β-adrenergic receptor agonist is a β2-adrenergic receptor agonist or a β3-adrenergic receptor agonists. Examples of β-adrenergic receptor agonists suitable for use with the methods of the present invention include, but are not limited to, albuterol, bitolterol, salbutamol, terbutaline, metaproterenol, procaterol, salmeterol, clenbuterol, isoproterenol, zinterol, BRL 37344, CL316243, CGP-12177A, GS 332, L-757793, L-760087, L-764646, and L-766892, etc. (see, e.g., Goodman and Gilman, supra).


[0077] Phosphodiesterase Inhibitors


[0078] In another preferred embodiment, the compound is a phosphodiesterase inhibitor. Cyclic nucleotide second messengers (cAMP and cGMP) play a central role in signal transductions and regulation of physiologic responses. Their intracellular levels are controlled by the complex superfamily of cyclic nucleotide phosphodiesterases (PDE) enzymes. Inhibitors of phosphodiesterases (PDE) are agents that can either activate or suppress PDEs via allosteric interaction with the enzymes or binding to the active site of the enzymes. The PDE family includes at least 19 different genes and at least 11 PDE isozyme families, with over 50 isozymes having been identified thus far. The PDEs are distinguished by (a) substrate specificity, i.e., cGMP-specific, cAMP-specific or nonspecific PDEs, (b) tissue, cellular or even sub-cellular distribution, and (c) regulation by distinct allosteric activators or inhibitors. PDE inhibitors include both nonspecific PDE inhibitors and specific PDE inhibitors (those that inhibit a single type of phosphodiesterase with little, if any, effect on any other type of phosphodiesterase). Still other useful PDE inhibitors are the dual selective PDE inhibitors (e.g., PDE III/IV inhibitors or PDE II/IV inhibitors). Below is a table setting forth various PDE inhibitors that are useful in the methods of the present invention.
2IsozymeRegulatorySelectiveFamilyCharacteristicsInhibitorsICa2+, Calmodulin-regulatedVinpocetinewith different Kmvalues for cG and cAhydrolysisIIICG-inhibited cAMilrinone,hydrolysis; low Km forAmrinone,cA and cGPimobendan,Cilostamide,Enoximone,Peroximone,VesarinoneIVLow Km for cA hydrolysisRolipram;RO-20-1724VHigh and low Km isoformsZaprinast;for cG specificDipyridamolehydrolysis


[0079] In one embodiment, the PDE inhibitor is a PDE V inhibitor. Useful phosphodiesterase type V inhibitors include, e.g., cialis, vardenafil , tanadafil, zaprinast, MBCQ, MY-5445, dipyridamole, furoyl and benzofuroyl pyrroloquinolones, 2-(2-Methylpyridin-4-yl)methyl-4-(3,4,5-trimethoxyphenyl)-8-(pyrimidin-2-yl)methoxy-1,2-dihydro-1-oxo-2,7-naphthyridine-3-carboxylic acid methyl ester hydrochloride (T-0156), T-1032 (methyl 2-(4-aminophenyl)-1,2-dihydro-1-oxo-7-(2-pyridylmethoxy)-4-(3,4,5-trimethoxy-phenyl)-3-isoquinoline carboxylate sulfate), and sildenafil. Cyclic GMP specific inhibitors include but not limited to A02131-1 [3-(5′-hydroxymethyl-2′-furyl)-1-benzyl thieno (3,2-c)pyrazole] for example. In another embodiment, the composition contains a phosphodiesterase type II (PDE II) inhibitor such as, e.g., EHNA. In yet another embodiment, the composition contains a phosphodiesterase type IV (PDE IV) inhibitor. Suitable phosphodiesterase type IV inhibitors include, but are not limited to, roflumilast, ariflo (SB207499), RP73401, CDP840, rolipram, mesopram, denbufylline, EMD 95832/3, cilomilast, RO-20-1724, and LAS31025. In still another embodiment, the phosphodiesterase inhibitor is a dual selective phosphodiesterase inhibitor such as, e.g., a PDE III/IV inhibitor (e.g., zardaverine) or phosphodiesterase inhibitors which can increase both cAMP and cGMP levels such as Satigrel (E5510, 4-cyano-5,5-bis(4-methoxyphenyl)-4-pentenoic acid).


[0080] In another embodiment, the PDE inhibitor is an inhibitor of the PDE III isozyme, for example, Olprinone.


[0081] In another embodiment, the PDE inhibitor is an inhibitor of the PDE IV isozyme family, or cAMP-specific and rolipram sensitive PDEs, which preferentially hydrolyze cAMP.


[0082] In yet another embodiment, the composition contains an agent that is a nonspecific phosphodiesterase inhibitor. Suitable nonspecific phosphodiesterase inhibitors include, but are not limited to, theobromine, dyphylline, IBMX, theophylline, aminophylline, pentoxifylline, papaverine, caffeine and other methylxanthine derivatives.


[0083] cAMP-Dependent Protein Kinase Activators


[0084] In other preferred embodiments, the compound used to treat the disorders described herein is a cAMP-dependent protein kinase activator. Examples of cAMP-dependet protein kinase activators include cAMP mimetics or dual cGMP/cAMP-dependent protein kinase activators. Suitable cAMP mimetics or analogs include those compounds that are structurally similar to cAMP and that have similar functions e.g., activities, as cAMP. Examples of suitable cAMP mimetics include, but are not limited to, 8-bromo-cAMP, dibutyryl-cAMP, Rp-cAMPS, and Sp-cAMPS, and useful dual activators include compounds such as, e.g., Sp-8-pCPT-cGMPS, Sp-8-bromo-cGMPS and 8-CPT-cAMP.


[0085] Superoxide Scavengers


[0086] In another embodiment, the compound used in the compositions and methods of the present invention is a superoxide anion (O2) scavenger. Superoxide can react with NO and dramatically reduce its biological effects. Accordingly, agents that scavenge superoxide anions can enhance the effects of NO. Examples of superoxide scavengers include, but are not limited to, exogenous Mn or Cu/Zn superoxide dismutase (SOD) or small molecule SOD mimetics such as, e.g., Mn(III) tetra(4-benzoic acid) porphyrin chloride (MnTBAP) and M40403 (see, e.g., Salvemini, et al., Science, 286(5438):304-306 (1999)).


[0087] Potassium Channel Activators


[0088] In another aspect, the present invention provides pharmaceutical compositions comprising a potassium channel activator. In one embodiment, the potassium channel activator is an ATP-sensitive potassium channel activator. Synthetic compounds that activate ATP-sensitive K channels are smooth muscle relaxants. Such compounds include, but are not limited to, minoxidil, minoxidil sulfate, pinocidil, diazoxide, levcromokalim, cromokalim, etc. (see, e.g., White, et al, Eur. J. Pharmacol., 357:41-51 (1998)). Additional suitable ATP-sensitive K channel activators can be found in, e.g., Bristol, et al., “Annual Reports in Medicinal Chemistry,” Vol. 29, Chap. 8, pp. 73-82, Academic Press (1991). In another embodiment, the potassium channel activator is a Maxi-K channel activator. Examples of activators of the Maxi-K channels include, but are not limited to, estrogen-like compounds, such as estradiol (see, Valverde, et al., SCIENCE, 285:1929-1931).


[0089] Benzodiazepines


[0090] In another aspect, the present invention provides pharmaceutical compositions comprising a benzodiazepine. Suitable benzodiazepines include, but are not limited to, alprazolam, brotizolam, chlordiazepoxide, clobazam, clonazepam, chlorazepate, demoxepam, diazepam, estazolam, flumazenil, flurazepam, halazepam, lorazepam, midazolam, nitrazepam, nordazepam, oxazepam, prazepam, quazepam, temazepam, and triazolam (see, e.g., Goodman and Gilman, supra).


[0091] Adrenergic Nerve Inhibitors


[0092] In another aspect, the compounds of the present invention are compounds that inhibit adrenergic nerves. Adrenergic nerve inhibitors include compounds that destroy sympathetic nerve terminals, such as 6-hydroxydopamine and its analogs (see, e.g., Goodman and Gilman, supra). Adrenergic nerve inhibitors also include compounds that deplete norepinephrine storage, either by inhibiting norepinephrine biosynthesis or be depleting stores, and compounds that inhibit norepinephrine release. Compounds that inhibit norepinephrine biosynthesis include, but are not limited to, α-methyltyrosine. Compounds that deplete norepinephrine stores include, but are not limited to, reserpine, guanethidine and bretylium. Compounds that inhibit norepinephrine release include, but are not limited to, clonidine and other (α2-adrenergic receptor antagonists. Examples of sympathetic nerve terminal destroyers include, but are not limited to, α2-adrenergic receptor antagonists.


[0093] Antidiarrheal Agents


[0094] In another aspect, the compounds of the present invention are antidiarrheal agents. Examples of suitable antidiarrheal agents include, but are not limited to, diphenoxylate, loperamide, bismuth subsalicylate, octreotide, etc. (see, e.g., Goodman and Gilman, supra).


[0095] HMG-CoA Reductase Inhibitors


[0096] In another aspect, the compounds of the present invention are HMG-CoA reductase inhibitors. Examples of HMG-CoA reductase inhibitors include, but are not limited to, mevastatin, lovastatin, simvastatin, pravastatin, cerivastatin, dalvastatin, atorvastatin, fluvastatin, etc. (see, e.g., Goodman and Gilman, supra).


[0097] Smooth Muscle Relaxants


[0098] In other embodiments, the compounds of the present invention are smooth muscle relaxants such as, e.g., hydralazine, papaverine, tiropramide, cyclandelate, isoxsuprine and nylidrin.


[0099] Adenosine Receptor Modulators


[0100] In another aspect, the present invention provides compositions for the treatment for a malignant cell phenotype comprising adenosine receptor modulators, either alone or in combination with another agent. Methods for the use of these compositions are also provided. In one group of embodiments, adenosine receptor modulators are used alone. In another group of embodiments, the adenosine receptor modulators are combined with at least one other muscle-relaxing agent. In other embodiments, the compounds of the present invention are adenosine receptor modulators such as methylxanthines. Examples of adenosine receptor modulators include theophylline and dyphylline. For other examples see, Goodman & Gilman, supra). Preferred agents are selected from those described with reference to the compositions of single agents or combinations above.


[0101] Theophylline, a plant-derived methylxanthine, has been used for the treatment of bronchial asthma for decades. Theophylline relaxes smooth muscle, notably bronchial muscle, that has been contracted experimentally with a spasmogen, or clinically in asthma. Proposed mechanisms of methylxanthine-induced physiologic and pharmacological effects include: 1) inhibition of phosphodiesterases, thereby increasing intracellular cyclic AMP, 2) direct effects on intracellular calcium concentration, 3) indirect effects on intracellular calcium concentrations via cell membrane hyperpolarization, 4) uncoupling of intracellular calcium increases with muscle contractile elements, and 5) antagonism of adenosine receptors.


[0102] A related compound, i.e., dyphylline, is a preferred adenosine receptor modulator. This compound is not metabolized by the liver and is excreted unchanged by the kidneys, therefore its pharmacokinetics and plasma levels are independent of factors that affect liver enzymes such as smoking, age, congestive heart failure, or the use of other drugs that affect liver function.


[0103] Adenylyl Cyclase Activators


[0104] In another aspect, the present invention provides compositions comprising adenylyl cyclase activators, either alone or in combination with other compounds or agents described herein. The adenylyl cyclase activator forskolin is preferred. Other examples of adenylyl cyclase activators, include, but are not limited to, N6, O2′-dibutyryl-cAMP, 8-chloro-cAMP, and Rp-diastereomers of adenosine 3′,5′-cyclic monophosphorothioate, and related analogs, such as Rp-8-bromo-adenosine 3′,5′-cyclic monophosphorothioate, and derivatives of forskolin, including colforsin daropate hydrochloride.


[0105] Endothelin Receptor Antagonists


[0106] In another aspect, the present invention provides compositions comprising endothelin receptor antagonist, either alone or in combination with other compounds disclosed herein. Examples of endothelin receptor antagonists include, but are not limited to, BE 1827B, JKC-301, JKC-302, BQ-610, W-7338A, IRL-1038, LRL-1620, bosetan, ABT 627, Ro 48-5695, Ro 61-1790, tesosentan (Ro 61-0612, ZD1611, BMS-187308, BMS-182874, BMS-193884, sitaxsentan (TBC 11251), TBC 2576, TBC 3214, TBC-10950, ABT-627, atrasentan, A-192621, A-308165, A-216546, CI-1020, EMD 122946, J-104132 (L753037), LU 127043, LU 135252, LU 302872, TAK-044 (69), SB 209670, SB 234551, SB 247083, ATZ1993, PABSA, L-749,329, RPR111723, RPR11801A, PD 164800, PD 180988 (CI1034), IRL 3630, IRL 2500, and their derivatives, etc. (see, Doherty, Annual Reports in Medicinal Chemistry, Volume 35, pp. 73-82, Academic Press, 2000). Other ethenesulfonamide derivatives, which are endothelin receptor antagonists and which are useful in the methods of the present invention, are disclosed by Harada, et al., Chem. Pharm. Bull., 49(12):1593-1603 (2001).


[0107] Bisphosphonates


[0108] In another aspect, the present invention provides compositions comprising bisphosphonates, either alone or in combination with other agents. Suitable bisphosphonates suitable for use in the methods of the present invention include, but are not limited to, alendronate sodium (Fosamax), pamidronate disodium (Aredia), etidronate disodium Ididronel) and the like.


[0109] cGMP-Dependent Protein Kinase Activators


[0110] In another aspect, the present invention provides cGMP-dependent protein kinase activators, either alone or in combination with other agents disclosed herein. Suitable cGMP-dependent protein kinase activators include, but are not limited to, cGMP mimetics or dual cGMP/cAMP-dependent protein kinase activators. Suitable cGMP mimetics or analogs include those compounds that are structurally similar to cGMP and that have similar functions, e.g., activities, as cGMP. Examples of suitable cGMP mimetics include, but are not limited to, 8-bromo-cGMP, dibutyryl-cGMP, Rp-cGMPS, and Sp-cGMPS, and useful dual activators include compounds such as, e.g., Sp-8-pCPT-cGMPS, Sp-8-bromo-cGMPS and 8-CPT-cAMP.


[0111] Guanylyl Cylase Activators


[0112] For example, BAY 41-2272 is a novel non-NO-based direct stimulator of soluble guanylyl cyclase that activates purified enzyme in a synergistic fashion with NO.


[0113] SOC Inhibitors


[0114] In another aspect, the present invention provides store-operated calcium influx (SOC) inhibitors, which inhibit calcium uptake into non-excitable cells in response to stimulus-mediated depletion of intracellular calcium storage pools. The SOC inhibitors preferably inhibit one or more of the following: calcium-dependent activation of nuclear factor of activated T cells (NFAT), nuclear factor kB (NF-kB), the stress kinase c-Jun N-terminal kinase (JNK) and exocytosis, resulting in the release or elaboration of inflammatory mediators. Examples of SOC inhibitors include for example statins in the δ-lactone form such as lovastatin, mevastatin, fluvastatin, pravastatin, dalvastatin, cerivastatin, atrovastatin and simvastatin.


[0115] Other treatment options include NO mimetics in combination with an antioxidant to further enhance efficacy. Such antioxidants include for example, lycopene, resveratrol, green tea polyphenolics (e.g. ECGC), brassinin (from cruciferous vegetables like Chinese cabbage), sulforaphane (from broccoli) and its analog sulforamate, withanolides (from tomatillos), and n-acetyl cysteine.


[0116] For purposes of the present invention by the term “animal” it is meant to include all mammals, and in particular humans. Preferably, NO mimetics are administered to an animal at risk for or suffering from a malignant cell phenotype. Such animals are also referred to herein as subjects or patients in need of treatment.


[0117] Low oxygen levels have been correlated with an increased level of cellular invasion and invasiveness. Hypoxic stress causes a variety of cellular adaptations, often manifesting in the up-regulation of certain genes.


[0118] For example, it has been shown that uPAR mRNA and cell surface uPAR protein levels increase under hypoxic conditions. uPAR is a high affinity cell surface receptor for pro-urokinase-type plasminogen activator (pro-uPA). Upon binding of pro-uPA to uPAR, the inactive single-chain pro-uPA is cleaved into its active, two-chain form. The activated enzyme, still attached to the receptor, then acts to convert plasminogen into plasmin, which ultimately degrades several components of the extracellular matrix (ECM). Active uPA also serves to activate both latent metalloproteinases and growth factors. uPAR also serves as a receptor for the ECM molecule vitronectin and can also modulate integrin function. In combination, these functions increase cellular invasion and potential for invasiveness. A positive correlation between hypoxia-induced uPAR up-regulation and carcinoma cell invasiveness has been suggested (Graham et al. Int. J. Cancer 1999 80:617-623). In addition, we have now shown hyponitroxia induced by administration of the nitric oxide synthase antagonist L-NMMA (0.5 mM) in hypoxic (1% O2) and nonhypoxic (5% and 20% O2) conditions to increase uPAR mRNA levels in human MDA-MD-231 cells incubated for 24 hours at 37° C.


[0119] Although the role of uPAR in invasion and tumor progression has been studied extensively, the regulatory mechanisms governing its expression are poorly understood. The invasive potential of a cell is highly dependent upon its ability to penetrate the extracellular matrix (ECM) and basement membranes that impede its movement. This process involves the participation of a number of proteolytic enzyme systems, of which the uPA system figures prominently. We postulate that the cGMP-dependent regulation of invasion and metastasis seen the current study was at least partially mediated through alterations in uPAR expression.


[0120] We suggest a novel mechanism of oxygen sensing and uPAR regulation whereby phenotypes are modified in response to a decrease in cGMP-dependent signaling. This phenomenon is due to a reduction in endogenous NO synthesis. Molecular oxygen is obligatory for the conversion of L-arginine into NO and L-citrulline by the enzyme NO synthase (NOS). Indeed, exposure of cells to low levels of oxygen (1-3%) inhibits NO production by up to 90% and NOS inhibition has been shown to induce invasive phenotypes in a manner similar to hypoxia. Due to the reduced NO levels, there is a decrease in GC activity and a consequential reduction in cellular cGMP. Indeed, Taylor et al. (1998) showed that culturing intestinal epithelial cells in hypoxia (1% O2; 24 Hrs) resulted in a significant decrease in basal and stimulated cGMP levels. Our studies have also suggested that a potential oxygen sensing mechanism of epithelial cells involves the participation of heme containing proteins such as GC. We have similarly shown that low oxygen levels decrease cGMP generation, and that they also result in phenotypic alterations. In both studies, these alterations were overcome by the addition of 8-Br-cGMP. We have further shown that inhibition of soluble guanylyl cyclase (sGC) with 1H-[1,2,4]Oxadiazole[4,3-a]quinoxalin-1-one (ODQ) can antagonize the ability of NO-mimetics to prevent the hypoxic up-regulation of uPAR. Given these findings we have derived a working model for the oxygen-mediated regulation of uPAR (FIG. 4). In this model, low oxygen levels result in decreased NO synthesis and a consequential decrease in sGC activation. As such, cellular cGMP levels are lowered, resulting in decreased PKG activity.


[0121] One possible mechanism involves a perturbation of the MAP kinase signaling pathway. This pathway is activated by hypoxia and studies have shown that NO can prevent the phosphorylation of ERK through a PKG-mediated interference of RAS/Raf. This concept has been strengthened by Yoshihide et al. (2000) who showed that NO donors and cGMP mimetics could reduce elastase levels by suppressing ERK phosphorylation. This led to a subsequent reduction in the activation and DNA binding capacity of AML1B (the transcription factor for elastase). It is possible that cGMP-dependent NO signaling similarly inhibits hypoxia-induced ERK phosphorylation, thereby decreasing the activation of the transcription factors responsible for the up-regulation of uPAR.


[0122] The promotor region of uPAR contains binding sites for transcription factors such as activator protein-1 (AP-1), Sp-1/3 and nuclear factor κB (NFκB). Hypoxia Inducible Factor 1 (HIF-1) levels may also contribute to the transcriptional activation of the uPAR gene, as previous examination of the sequence upstream of the uPAR initiation codon revealed the presence of at least 3 potential HIF-1-binding sites. It has also been shown that HIF-1 accumulation and transcriptional activity can be reduced by relatively high concentrations (2.5-500 μM) of NO mimetics such as SNP, S-nitroso-L-glutathione and 3-morpholinosydnonimine. Preliminary studies in our laboratory have confirmed that high concentrations (0.1-1 mM) of GTN and SNP inhibit HIF-1 accumulation and transactivating activity. However, concentrations of the NO mimetics that completely inhibited the hypoxic stimulation of metastasis, invasiveness and uPAR expression had no effect on HIF-1 accumulation or transactivating activity under hypoxia. Similarly, the administration of 8-Br-cGMP did not affect HIF-1 activity. Therefore it is unlikely that the cGMP-dependent regulation of invasive phenotypes is mediated via alterations in HIF-1.


[0123] Like uPAR, PAI-1 has also been shown to be stimulated under hypoxic conditions. See WO99/57306. Further, this stimulation was accompanied by a decrease in cellular adherence. PAI-1 is 52-kDa ECM glycoprotein which is produced by a variety of normal and malignant cells. This glycoprotein is a regulator of plasminogen activator activity. It functions to inhibit both free and bound uPA through the formation of irreversible covalent complexes. PAI-1 has also been shown to compete with the uPAR for binding to the same domain of vitronectin. As such, PAI-1 is capable of releasing cells bound to vitronectin-coated plates. Studies have shown that PAI-1 is required for the optimal in vitro invasiveness of lung carcinoma cells.


[0124] Hypoxia has also been shown to increase the resistance of cells to cytotoxic agents. The gene for PROXY-1 was identified using an RT-PCR based differential display following the culture of a variety of cell types under low levels of oxygen. See WO99/57306. It is believed that the 43-kDa PROXY-1 protein plays a role in protecting cells from insults including hypoxia, DNA damaging agents, cytotoxic agents and glucose deprivation, as enhanced PROXY-1 expression is observed in response to each of these harmful stimuli. Together with the fact that this gene is expressed by a variety of unrelated cell types, this type of gene expression is indicative of PROXY-1 being a universal ‘switch’ involved in the initial events that lead to cellular adaptations to hypoxia.


[0125] However, it is possible that the role of NO in tumor cell initiation promotion, progression, survival and apoptosis depends on the cell types, the concentrations of NO in the given cellular microenvironment, time of cellular exposure to NO, and possibly other factors. A similar paradigm has been drawn from the role of NO in acute and chronic inflammatory processes and in myocardial and neuro-protective preconditioning, for example. As a plieotropic transcriptional factor, NFκB (nuclear factor kappa B)-induced antiapoptotic signals have been suggested to be responsible for the development of chemoresistance in various forms of cancer, in tumor progression, and in the development of radiation resistance. NF-κB as a molecular target for developing anti-cancer therapy has been reviewed extensively by several investigators. Various known NFκB inhibitors such as NSAIDs, glucocorticoids, COX 2 inhibitors, and more recently proteasome inhibitors (blocks NF-κB activation), have been shown to be effective or potentially effective cancer treatment options. Known NFκB inducible genes that may be involved in tumor progression and chemoresistance include VEGF (vascular endothelium growth factor), EGFR (epidermal growth factor receptor), COX2 (cyclooxygenase type 2), MMPs (matrix metalloproteases 2 and 9 for example), uPAR (urokinase plasminogen activator receptor), etc. In a recent review article on inflammation and cancer, anti-inflammatory therapy is considered an efficacious approach towards early neoplastic progression and malignant conversion


[0126] It is possible that NO may function as the feedback inhibitor of NFκB upregulation/activation. Alternatively, NO may affect p53 tumor suppressor gene expression, or BcL expression. It is also possible that NO may enhance chemosensitivity of tumor cells via a completely unknown mechanism. While the traditional approach to treat cancer with combination chemotherapeutic agents and/or radiation therapy is primarily based on the known mechanism of action, current approach focuses on the balancing the effectiveness of the treatment.and toxicity profile of selected drugs. Since many of the NO mimetics are proven to be safe for both acute and chronic usage, it presents as a unique opportunity to minimize the safety risk when NO mimetics is added to the known standard of treatments of various forms of cancer.


[0127] In some forms of cancer, for example, prostate and breast cancers, hormonal therapy is typically used for early phase of the diseases. Hormonal therapy includes anti-androgens (e.g. flutamide) for prostate cancer and anti-estrogen (e.g. tamoxifen) for estrogen receptor positive or ER status unknown early stage breast cancers. In general, the tumor responded well to the hormonal therapy before a new phenotype is developed. The newly developed phenotypes are typically no longer responding to the original hormonal therapy; prognosis for patients at this stage of cancer, i.e. hormone refractory/insensitive phase is typically very poor. Even while using hormonal therapy during the hormone responding phase of cancers, patients suffer from hot flashes, loss of libido, sexual dysfunction, osteopenia, osteoporosis, poor self-esteem and quality of life. The mechanism leading to the loss of hormone response of these cancer phenotypes are largely unknown at this point. It could be related to androgen/estrogen receptor expression levels, cellular locations, functional activities of these steroid receptors. Alternatively, it could also due to the altered non-genomic actions of these steroid targets at various cellular compartments and sub-compartment, e.g. mitochondria.


[0128] Endogenous androgens including testosterone and 5-α-dihydrotestosterone (DHT; one of the two key metabolites of testosterone) are essential for the development and maintenance of reproductive tissues (e.g. testis, epididymis, seminal vesicles, penis, etc.) in male species. Exogenously administered androgens, e.g. testosterone undecanoate, testosterone propionate, etc. typically transformed to testosterone or DHT before exerting its biological activity. The majority of the biological activities of testosterone and DHT are elicited by the cytosolic androgen receptor (AR). The AR is a member of the steroid hormone-thyroid hormone-retinoic acid nuclear receptor super-family. Upon androgen binding, the AR undergoes conformational changes resulting in the release of inhibitory proteins and is subsequently hyper-phosphorylated, translated to the cell nucleus. Once in the nucleus, AR is dimerized and binds to hormone response elements in the regulatory regions of androgen target genes, and subsequent gene transcriptional events. DHT binds to AR with higher affinity and induces a higher level of androgen-regulated gene expression than testosterone. Since the second key metabolite is E2, the biological activity of testosterone is also mediated at least in part, by the activation of estrogen receptor.


[0129] Evidence has been recently obtained for ligand-independent activation of AR transcriptional activity by peptide growth factors, such as IGF-I (insulin-like growth factor), and to a lesser extent, keratinocyte growth factor and epidermal growth factor. Like other steroid receptor ligands, testosterone can also elicit biological activities via non-genomic mechanisms that often involve ion fluxes in androgen-dependent cell types. Specifically, calcium fluxes following addition of picomolar concentrations of testosterone have been reported in rat heart myocytes, male rat osteoblasts, human prostate cancer cells, and Sertoli cells. Direct evidence from patch-clamp studies showed that testosterone can open K+-channels in single coronary myocytes. Although testosterone mediated ion fluxes have been reported in various cell types, the biological consequences of these activities are still poorly defined.


[0130] While infrequent in early and localized prostate tumors, mutations of the AR have been identified in a number of advanced prostatic cancers. Substantial evidence suggests that AR activity is enhanced in advanced forms of prostate cancer, and in some cases, as a result of gene amplification events. Frequently, the AR also appears to have altered ligand specificity; for example, advanced prostate cancers often become androgen-independent, and no longer respond to androgen ablation therapy. The loss of androgen-sensitivity is considered an indicator of poor prognosis for advanced prostate cancers.


[0131] NO mimetics may be able to maintain the tumor cell under homeostatic stage, thus prevent the alteration of steroid receptor expressions, levels of expression, location, or alternatively, the ion-channels related to androgen and anti-androgen action etc. Thus, the administration of NO mimetics could keep patients under hormonal therapy for various forms of cancer under hormone responsive phase, preventing or delaying metastasis to secondary sites and transformation to more advanced, hormone-refractory/insensitive/insensitive cancers. NO mimetics can be administered in conjunction with hormonal therapy during the treatment phase and/or can be used in the remission phase.


[0132] In certain cases, NO mimetics can be used with chemo-and/or radio-therapeutic treatment to ensure eradication of cancers during the treatment phase, and can be used as stand-alone therapy during the remission. If low dose of chemotherapy is employed to prevent cancer recurrence, NO mimetics can be used with the low dose chemotherapy to prevent or prolong the time to cancer recurrence and eventually, prolong the survival time of cancer patients. The addition of NO mimetic therapy could also 1) reduce the dependence on narcotic pain relief agents and thus associated adverse events due to their use and yet enhance the effectiveness of these agents, 2) prevent progressive lost of bone mineral density and thus, the development of osteoporosis and the risk of bone fracture, and 3) improve overall quality of life of cancer suffers.


[0133] We have now found that nitric oxide is a primary mediator of cellular adaptive responses to changes in oxygen levels in and around the cell. Through administration of a low dose of a nitric oxide mimetic, we have shown that nitric oxide mimetic activity can be increased, restored or maintained at a level which inhibits or prevents a malignant cell phenotype. In contrast, the effect of maintaining low oxygen levels on cells was limited to inhibiting basal levels of endogenous nitric oxide production.


[0134] Under hypoxic conditions where.the levels of oxygen are limiting, we have now demonstrated that cancer cell lines acquire one or more of the following malignant cell phenotypic properties: they increase their lung-colonization ability following i.v. inoculation into syngeneic mice (experimental metastasis); they increase their invasiveness through the extracellular matrix in vitro (also relevant to metastasis); and they become more resistant to the chemotherapeutic drug doxorubicin and 5-fluorouracil. In these experiments, cancer cells were exposed to 1% O2 (10-15 mmHg PO2) to induce hypoxia and compared with nonhypoxic cancer cells exposed to 5-20% O2 (30-160 mmHg). By administering low doses of nitric oxide mimetics (during periods when oxygen levels are limiting and/or when endogenous nitric oxide production is inhibited) acquisition of these malignant phenotypic changes is prevented. This prevention occurs even when the cells are in an extremely hypoxic environment.


[0135] We have now demonstrated that low concentrations of nitric oxide mimetics SNP and/or glyceryl trinitrate (GTN) inhibit the hypoxic up-regulation of uPAR and PAI-1, as well as PROXY-1. Similar low dose nitric oxide mimetic therapy is expected to also be effective in inhibiting hyponitroxic upregulation of these genes such as that observed in cells treated with L-NMMA (0.5 mM), i.e. inhibiting and preventing a malignant cell phenotype.


[0136] Experiments performed in human breast cancer cells cultured in hypoxic conditions (1% O2) showed that treatment of the hypoxic cells with 10−12 M SNP significantly reduced levels of uPAR MRNA as compared to untreated control hypoxic cells and hypoxic cells treated with 10−8 M SNP. Similarly, treatment of breast cancer cells cultured in hypoxic conditions with the nitric oxide mimetic GTN at low doses of 10−11 M and 10−10 M significantly reduced uPAR mRNA levels in the hypoxic cells as compared to untreated hypoxic cells and to hypoxic cells treated with GTN at 10−9 M, 10−8 M, 10−7 M, 10−6 M and 10−5 M. In fact, levels of uPAR mRNA in hypoxic breast cancer cells treated with 10−11 M GTN and 10−10 M GTN were similar to levels of uPAR mRNA measured in cells cultured under non-hypoxic conditions (20% O2). Levels of uPAR mRNA in hypoxic cells treated with GTN at 10−6 M and 10−5 M were similar to levels of untreated hypoxic cells, suggesting tolerance to the NO mimetic. A reduction in uPAR protein levels was also observed in these cells 24 hours after incubation with these nitric oxide mimetics.


[0137] Further experiments with human invasive trophoblast cells (HTR-8/SVneo) confirmed the ability of low doses of these nitric oxide mimetics to decrease expression of uPAR in hypoxic cells.


[0138] The effects of treatment of HTR-8/SVneo invasive trophoblast cells with the nitric oxide mimetic GTN on PROXY-1 mRNA levels was also examined. PROXY-1 mRNA levels were very low in cells cultured in 20% O2. However, levels of PROXY-1 mRNA were increased in cells cultured in 1% O2 which were untreated or treated with 10−7 M GTN. In comparison, levels of PROXY-1 were much lower in hypoxic cells treated with a low dose, 10−11 M GTN.


[0139] In addition, the effects of the nitric oxide mimetics SNP and GTN on PAI-1 mRNA levels in breast cancer cells were examined. Again, treatment of hypoxic cells with low doses of the nitric oxide mimetics SNP (10−12 M) and GTN (10−11 M) significantly decreased levels of PAI-1 mRNA as compared to untreated hypoxic cells.


[0140] Experiments were also performed to ascertain the effects of low doses of nitric oxide mimetics on levels of metalloproteinase. Breast cancer cells were incubated in hypoxic or control conditions in the presence of varying concentrations of SNP or GTN. Treatment of hypoxic cells with low doses, 10−11 M GTN and 10−12 M SNP, of a nitric oxide mimetic resulted in a decrease in gelatinases secreted from the cells as compared to untreated hypoxic cells.


[0141] Functionally, inhibition of the hypoxic up-regulation of these genes was then shown to result in a decrease in cellular invasiveness and drug resistance. The invasive ability of cells in hypoxic conditions in the presence or absence of nitric oxide mimetics was also assessed using Matrigel invasion chambers (modified Boyden chambers). In these in vitro invasion assays, either breast cancer cells (see FIG. 1) or HTR-8/SVneo invasive trophoblasts were plated on Matrigel-coated membranes. Cells were then incubated under hypoxic or normal conditions, alone or in the presence of nitric oxide mimetics. The invasion index for each treatment was determined by staining the cells which invaded through the membrane and counting them. In both cell lines, treatment with low doses of nitric oxide mimetics significantly reduced hypoxic cell invasiveness as compared to untreated hypoxic cells. The invasive indices of hypoxic breast cancer cells treated with 10−10 M SNP and 10−11 M GTN were similar to or even lower than cells cultured under non-hypoxic conditions. In HTR-8/SVneo trophoblast cells, 10−7 M GTN inhibited invasiveness of hypoxic cells by 56.2%, while 10−8 M SNP inhibited invasiveness by 63.4%.


[0142] The ability of low doses of nitric oxide mimetics to inhibit metastases of tumor cells in animals was then confirmed. In a first set of experiments, the ability of hypoxic conditions to increase number of metastases was demonstrated. In these experiments, mice were administered via tail vein injection a bolus of metastatic melanoma cells. Immediately after injection the mice were divided into two groups. The first group was placed in a chamber with a continuous flow of a gas mixture comprising 21% O2 (room air). The second group was placed in a hypoxic environment with only 10% O2. After 24 hours, both groups were removed and placed in regular cages kept at room air. After 14 days, the animals were sacrificed and metastatic nodules in the lungs of the animals were counted. Animals in the hypoxic environment had a 2-fold statistically higher number of lung nodules as compared to animals in the non-hypoxic environment.


[0143] In a second set of experiments, mice were injected with mouse melanoma cells which were pre-incubated for 12 hours in 1% or 20% O2 in the presence or absence of a low concentration of a nitric oxide mimetic (GTN; 2×10−11 M). After fourteen days, the mice were sacrificed and the lungs were visually observed for metastatic nodules. In addition, the number of lung nodules in these mice was compared. Lungs of animals that had been administered hypoxic and non-hypoxic melanoma cells treated with the nitric oxide mimetic prior to injection exhibited statistically less metastatic nodules as compared to animals administered either untreated hypoxic and non-hypoxic melanoma cells (see FIG. 2). Specifically, the in vitro pre-treatment with 2×10−11M GTN decreased the hypoxia-stimulated lung nodule formation by 85% and, even in 20% oxygen, the GTN pre-treatment reduced the extent of metastasis by more than 60%. In fact, the suppression of lung nodule formation by GTN pre-treatment was found to be equivalent regardless of the in vitro oxygenation levels. Further, treatment of the cells with L-NMMA prior to inoculation into mice resulted in a 63% overall increase in the number of lung nodules (p<0.01; post hoc Fisher's test). Concomitant treatment using GTN (10−10M) and L-NMMA attenuated this metastatic response by 60% (p<0.0005). The frequency distribution of lung nodules across the three experimental groups ranged from 0 to 111. No lung nodules were found in two of the control mice and four of the GTN-treated mice. Characterization of the lung nodule frequency by tertile revealed a consistent pattern of suppression throughout the NO-mimetic treated group compared to the L-NMMA treated group. In addition, the metastasis in the GTN-treated group was significantly below even the control levels in the highest tertile. Taken together, these data indicate that the levels of NO, and not oxygen itself, determine the severity of the metastatic phenotype. Further, the effect of low-concentration GTN treatment on lung nodule formation was not due to a non-specific cytotoxic or growth inhibitory effect on the cells as they had similar in vitro colony-forming ability as untreated cells.


[0144] As will be understood by those skilled in the art upon reading this disclosure, results from the studies in this murine model can be used to predict drug disposition in other species including humans, to define pharmacokinetic equivalence in various species including humans and design dosing regimes for other experimental animal models and for human clinical studies. Such pharmacokinetic scaling is performed routinely based upon data such as provided herein as evidenced by references such as Mordenti, J. J. Pharm. Sci. 1986 75(11):1028-1040.


[0145] Further, the ability of an NO mimetic to reduce disease progression in humans was demonstrated. In this study, continuous transdermal patches were used to deliver very low doses of GTN (0.033 mg/hour) to patients with recurrent prostatic adenocarcinoma. Patients with prostatic adenocarcinoma were selected for this study because the progression of this type of cancer correlates well with the plasma levels of prostate-specific antigen (PSA). Thus, the outcome of low dose NO mimetic therapy can be easily assessed by measuring PSA levels. Analysis of data from two patients in this study revealed a sharp decline in plasma PSA levels within two months of GTN treatment, thus indicating low dose NO mimetic therapy to be an effective approach to the management of cancer, particularly prostate cancer, in humans (see FIGS. 3A and 3B). Plasma PSA levels were measured via a commercially available immunoassay kit such as Immuno 1 (Bayer Corporation).


[0146] The ability of low doses of nitric oxide mimetics to decrease resistance of breast cancer cells to doxorubicin was also examined. In these experiments, the ability of hypoxic conditions to increase resistance to doxorubicin was first confirmed. Cells exposed to 1% O2 had higher survival rates at concentrations of 25 and 50 μM doxorubicin as compared to cells exposed to 20% O2. The effect of low doses of the nitric oxide mimetic GTN on doxorubicin resistance of hypoxic and non-hypoxic cells was then examined. It was found that hypoxic cancer cells treated with 10−6 M and 10−10 M GTN had lower doxorubicin survival rates as compared to untreated hypoxic cells. The survival rates of the nitric oxide mimetic treated hypoxic cells were comparable to those observed in untreated non-hypoxic cells and non-hypoxic cells treated with the nitric oxide mimetic. These results have been confirmed in multiple human cancers as well as mouse cancers and with other antimalignant therapeutic modalities.


[0147] Thus, these studies demonstrate that a malignant cell phenotype such as that induced by hypoxia can be inhibited and prevented by increasing (restoring) the level of nitric oxide mimetic activity. Other factors known to lower cellular nitric oxide mimetic activity so as to induce a malignant cell phenotype include, but are not limited to, decreases in arginine levels, exposure to endogenous nitric oxide synthase antagonists such as L-NMMA and ADMA, exposure to endogenous nitric oxide scavengers such as superoxide, changes in nitric oxide synthase expression, changes in cofactors such as GSH and NADPH, glucose deprivation, surgical procedures, administration of anesthetic agents, administration of pharmacologic agents which alter circulation such as, but not limited to antihypertensive agents, and traumatic injuries including, but not limited to those associated with blood loss, decreased blood volume, and hemorrhage. The present invention relates to methods of inhibiting and preventing a malignant cell phenotype resulting from these and other factors by administering low doses of one or more nitric oxide mimetics.


[0148] The low dose nitric oxide mimetic therapy of the present invention will also prevent the malignant cell phenotype of vascular endothelial cells which ultimately results in recruitment of such cells by a tumor and development of a blood supply to the tumor, also known as angiogenesis. Attempts at cutting off tumor blood supply by blocking VEGF in vascular endothelial cells have been relatively unsuccessful as cancer treatments. The presence of VEGF in tumors has been shown to suppress tumor invasiveness. Thus, it is believed that agents which remove or block the actions of VEGF, such as anti-VEGF antibody, actually generate more aggressive cancer cell phenotypes. Using the present invention, however, generation of a more aggressive malignant cell phenotype can be prevented, thereby allowing use of anti-VEGF therapies to prevent angiogenesis without producing more aggressive cancer cells.


[0149] These studies also demonstrate the ability of low dose NO mimetic therapy to decrease levels of a tumor marker in a patient. Tumor marker levels are used routinely by those of skill in the art as indicators of the progression of a cancer. Accordingly, the ability of low dose NO mimetic therapy to decrease levels of these markers is indicative of its ability to treat cancers. Further, progression of a tumor can be diagnosed and/or monitored in a patient by measuring the levels of a tumor marker in the patient in the presence of a low dose of a nitric oxide mimetic.


[0150] Low dose formulations of nitric oxide mimetics which ultimately result in an increase, restoration or maintenance of nitric oxide mimetic activity of cells sufficient to prevent or inhibit a malignant cell phenotype can be produced in accordance with formulation methods known in the art. Formulations for the administration of nitric oxide mimetics in accordance with the method of the present invention can take the form of ointments, transdermal patches, transbuccal patches, injectables, nasal inhalant forms, spray forms for deep lung delivery through the mouth, orally administered ingestible tablets and capsules, and tablets or lozenges, or “lollipop” formulations for administration through the oral mucosal tissue. The latter formulations included tablets, lozenges and the like which are dissolved while being held on or under the tongue, or in the buccal pouch. It is preferred that the pharmaceutical preparations provide a low dose of the nitric oxide mimetic sufficient to increase, restore or maintain nitric oxide mimetic activity at a level which inhibits or prevents a malignant cell phenotype, also referred to herein as a therapeutically effective amount, during the period in which cellular nitric oxide mimetic activity of cells is lowered. Also preferred are formulations comprising more than one NO mimetic. In this embodiment, it is preferred that the NO mimetics target or act on different parts of the NO pathway. For example, an NO donor can be co-administered with a compound that inhibits cyclic nucleotide (e.g. cAMP or cGMP) degradation such as a phosphodiesterase inhibitor. Preferred phosphodiesterase (PDE) inhibitors useful as NO mimetics are those inhibiting PDE-1 through PDE-5.


[0151] While the methods of the present invention are useful for treating a broad range of diseases, they are particularly effective for lung cancer, including non-small cell lung cancer and small cell lung cancer; mesothelioma; thymic tumors; breast cancer, such as ductal carcinoma in situ, lobular carcinoma in situ, ductal carcinoma of special type, and invasive lobular carcinoma; colorectal cancer; anal cancer; esophageal cancer; gastric cancer; small intestine and carcinoid tumors; cancers of the liver such as primary liver cancer and hepatocellular carcinoma; cancer of the biliary tract, including cholangiocarcinomas; tumors of the gall bladder; pancreatic cancer; endocrine cancers such as thyroid cancer, adrenal cancer, and neuroblastoma; genitourinary cancers such as renal cancer, Wilms' tumor, cancer of the bladder and ureter, prostate cancer, and testicular cancer; gynecological cancers such as ovarian cancer, cancer of the uterine corpus, cancer of the cervix, vaginal and vulvar cancer, and trophoblastic tumors; head and neck cancer such as cancer of the larynx, cancer of the oral cavity, carcinoma of the nasopharynx, cancer of the nasal cavity and paranasal sinuses, cancer of the eye and orbit, and salivary gland tumors; tumors of the central nervous system such as primary brain tumors and brain metastatses; skin cancer, including primary cutaneous malignant melanoma and non-melanoma skin cancers; hematological malignancies including acute leukemia, chronic lymphoid leukemias, Hodgkin's disease, non-Hodgkin's lymphomas, and myeloma; bone and soft tissue malignancies such as osteosarcoma, Ewing's sarcoma, other primary bone tumors, and soft tissue sarcomas; cancer of unknown primary site; paraneoplastic syndromes, including endocrine paraneoplastic syndromes, neurological paraneoplastic syndromes, hematological paraneoplastic syndromes, and dermatological paraneoplastic syndromes; and AIDS-related malignancies, including Kaposi's sarcoma, non-Hodgkin's lymphoma, and primary central nervous system lymphoma. Table 2 provides an exemplary list of cancers which can be treated using the methods and formulations of the present invention, and the current standard of care used to treat these cancers as well as investigational treatments used to treat these cancers.
3TABLE 2Standard of Care andType of CancerDescription of the CancerInvestigational TreatmentsLung CancerLung cancers are grouped asSurgical removal of non-small cell bronchogenicnon-small cell (NSCLC) orcarcinoma continues to offer best possibility of asmall cell (SCLC), butcure.within the former certainRadical radiotherapy is indicated for patients withpatterns of disease do relatewho are unfit for surgery, or have goodto histological subtype. Forperformance status and disease that can beexample, squamous cancersencompassed in a radical volume. The standardtypically arise in proximalinternational dose is 60 Gy in 30 fractions over 6segmental bronchi and growweeks. The standard treatment volume in most ofslowly, disseminatingthe world is the primary tumor and hilar andrelatively late in their course.mediastinal lymph nodes, with a 1-2 cm margin.Adenocarcinomas are oftenChemotherapy delivered synchronously withperipheral in origin and evenradiotherapy is being investigated.small respectable lesionsCisplatin-based therapy significantly improvescarry a risk of occultsurvival and quality of life. The most studiedmetastases.regimen is MIC (mitomycin C, isosfamide, andcisplatin)..Combination chemotherapy is now the standardtreatment for limited and extensive SCLC.Patients with small cell lung cancer (SCLC) areusually treated by primary chemotherapy becauseof its chemo-responsiveness and frequentdissemination at time of diagnosis. SCLC is,however, also the most radio-responsive variety ofbronchial carcinoma and radiotherapy has animportant role in its management. In patients witha localized tumor, thoracic irradiation (TI) andprophylactic cranial irradiation (PCI) improvedisease control at these sites and can lead toprolongation of survival.The following courses of radiotherapy arecommonly used: TI-50 Gy in 2Gy daily fractionsor biological equivalents; PCI-24 Gy in 8fractions, 30 Gy in 10 fractions, or 36 Gy in 18fractions. It is common practice to irradiate themediastinum even in the absence oflymphadenopathy because of frequency of occultnodal metastases.A number of chemotherapeutic drugs have beenfound to have activity as single agents and avariety of combination regimens have beendeveloped. CAV (cyclophosphamide, doxorubicinand vincristine) and EP (etoposide and cisplatin)have similar efficacy.Malignant pleuralAggressive tumor arisingWithout treatment, the average patient with MPMmesothelioma (MPM)from the serosal lining of thesurvives less than one year from the time ofchest and abdomen withdiagnosis. As a result of this poor prognosissurvival rates of less thanvarious therapeutic options have been explored,one year reported followingincluding radiotherapy, chemotherapy,diagnosis.immunotherapy and surgery. Current experiencehas shown multimodality therapy to offerimproved survival in selected patient groups.Thymic TumorsTumors derived from theThe contemporary standard of treatment forthymus (thymomas)thymoma is radical surgery whenever possible.comprise approximately 20%Total resection alone is adequate therapy for earlyof all mediastinal tumors andstage thymoma. Adjuvant therapy should be usedare the most common tumorfor local disease with demonstration or suspicionin the anderior mediastinum.of invasion. In local disease with or withoutThymomas occur at any ageresidual tumor, radiotherapy is recommended inbut are rare before the age ofdoses of 50-60 Gy given in 20-3020 and peak between 40-60fractions. Locally advanced, relapsing, oryears.disseminated disease should be treated withMost thymomas are slow-combination chemotherapy includinggrowing ‘low-grade’corticosteroids. Cisplatin-based regimens aremalignant tumors. It isrecommended.believed that they derivefrom epithelial elements, butthe tumors retain thecapacity for production of Tcells.Breast CancerBreast cancer is the mostEarly breast cancer is defined as disease that cancommon solid cancerbe completely extirpated by surgery. Theoccurring in women. It ismanagement of this disease comprises: treatmentestimated that 5-10% ofof the breast and axilla; pathological staging tofemale breast cancer is duedirect adjuvant therapy; adjuvant therapy-to inheritance of a mutatedendocrine, chemotherapy, radiotherapy; andcopy of either BRCA1 orfollow-up.BRCA2. Other genescontribute less frequently toAll patients require removal of the primary tumorfamilial breast cancer. Malewith either wide local excision or mastectomy.breast cancer is rare (0.7% ofBreast irradiation has been shown to reduce theall male cancers) with a peakrisk of local recurrence after breast-conservingincidence 10 years later thansurgery. Typically, the whole breast is treatedwomen. It may occur inwith tangential fields to a dose of 50 Gy in 25association with Klinefelter'sfractions (or an equivalent dose-fractionationsyndrome.regimen), with care taken to minimize the volumeDuctal carcinoma in situof lung and heart irradiated.(DCIS)Primary chemotherapy or hormone therapy for90% of breast carcinomasoperable breast cancer provides early systemicarise in the ducts of thetreatment and allows assessment of the responsebreast, beginning as atypicalto treatment; by definition this is impossible withproliferations of ductaladjuvant therapy. Studies have compared pre- andepithelium that eventuallypost-operative chemotherapy (doxorubicin andfill and plug the ducts withcyclophosphamide). Pre-operative treatment doesneoplastic cells. As long asdownstage the primary tumor and may facilitatethe tumor remains within thebreast-conserving surgery where mastectomyconfines of the ductalwould otherwise be required.basement membrane it isLocally advanced disease is defined by theclassified as DCIS. Not allpresence of infiltration of the skin or the chestDCIS will inevitablywall or fixed auxillary nodes. The probabilityprogress, but the probabilityof metastatic disease is high.of development of invasivecancer is estimated at 30-50%.Lobular carcinoma in situLocal control of the tumor and the prevention ofThese pre-invasive lesionsfungation are of major importance to the qualitycarry a risk not only ofof life. A combination of primary systemicipsilateral invasive lobulartreatment and radiotherapy is commonly used.carcinoma but also ofMany of these patients have indolent ER-positivecontralateral breast cancer.disease that responds to endocrine therapy withInvasive ductal carcinomatamoxifen, aromatase inhibitors, or progestins. InThis accounts for 75% ofyounger women with aggressive ‘inflammatory’breast cancers. Thebreast cancer, primary chemotherapy is preferred.malignant cells areIn patients with a good response to systemicassociated with a fibroustreatment surgery may be feasible.stroma that can be denseIn management of metastatic breast cancer,(scirrhous carcinoma). Thetreatment with tamoxifen, ovarian ablation,tumor invades throughprogestins, or aromatase inhibitors may providebreast tissue into thean response in patients with advanced disease,lymphatics and vascularand in 50-60% of those with ER-positive tumors.spaces, to gain access to theDisease that responds to endocrine therapy andregional nodes (auxiliarythen progresses has a 25% response rate withand, less often, internalsecond-line treatment; the response to a thirdmammary) and the systemicagent is 10-15%.circulation.Advanced breast cancer is moderatelyDuctal carcinoma of specialchemosensitive. Active agents includetypeantracyclines, alkylating agents, andA number of pathologicalantimetabolites. Combinations such as FAC (5-variants are identified,fluorouracil, doxorubicin, cyclophosphamide)namely medullaryproduce response rates of 40-60%, with a mediancarcinoma, tubulartime to progression of around 8 months. 20-50%carcinoma, and mucinousof women respond to second-line chemotherapycarcinoma.with a taxane and studies are currently evaluatingInvasive lobular carcinomathe promising combination of anthracycline plusLobular carcinomas accounttaxane as first-line chemotherapy. Low-dosefor 5-10% of breast cancers.radiotherapy provides palliation in these patients.Bisphosphonates are used for treatment ofmalignant hypercalcemia and can produce healingof some osteolytic metastases.Colorectal cancerColorectal cancer is theSurgery is the mainstay of curative therapy forfourth commonest cancercolorectal cancer. Curative resection requires theworldwide. Around two-excision of the primary tumor and its lymphaticthirds of a million peopledrainage with an enveloping margin of normalwill present with the diseasetissue.each year. It affects men andThe aim of adjuvant chemotherapy is to eradicatewomen almost equally andmicro-metastases and thereby prevent futuretends to be more common inrelapse. Current available chemotherapy does not‘developed’ countries and iscompletely eradicate bulky, advanced metastaticparticularly common in thebowel cancer. It does however eradicate mico-US, Europe and Australia.metastases in a proportion of patients.Colorectal cancers areaThe current ‘international standard’ adjuvantalmost always andenocarcinomas. Thechemotherapy regimen for colon carcinoma is 5-tumor often starts as afluorouracil (5FU) given in combination withpolypoidal mass and thenfolinic acid, by bolus intravenous injection.tends to infiltrate into andthrough the bowel wall.A number of new oral preparations involving pro-drugs of 5FU and/or inhibitors of 5FU catabolismare being evaluated. Other research approachesinvolve regional delivery of 5FU to the tissuesmost at risk of containing micro-metastases, viathe peritoneal cavity or the hepatic portal vein.New cytotoxic agents are continually underinvestigation, the most promising of which areIrinotecan (a drug which works by interfering withthe nuclear enzyme topoisomerase-I), andOxaliplatin (which forms covalently bondedadducts with DNA).Understanding the complex pathways by which5FU acts has allowed the development ofbiochemical modulation techniques. Leurovorinenhances 5FU toxicity by stabilizing theinteraction of thymidylate synthase and active5FU metabolites. The optimum schedule of 5FU ±leucovorin administration has yet to be elucidated.Intravenous bolus 5FU, usually given withleucovorin, is currently most commonly used.Raltitrexed is a potent inhibitor of thymidylatesynthase. It can be used as an alternative to 5FU-based regimes for first-line treatment. It does notrequire coadministration of a biochemicalmodulator. Intra-hepatic 5FU, following surgicalplacement of a hepatic artery catheter, may beconsidered for patients with unresectable livermetastases.There currently is no standard second-linechemotherapy treatment regime for patientspreviously treated with 5FU-based chemotherapy.Radiotherapy in colonic cancer is limited to thepalliative situation in most circumstances. Therectum is a suitable target for radiotherapy.Radiotherapy has been used in both the pre-operative and post-operative settings in thisdisease. In the pre-operative situation there are agroup of patients who present with large fixed ortethered tumors that are non-resectable. Theconversion rate to respectability is 35-75%, with adose of 50-60 Gy given over a five-week period.This group is also offered combined chemo-irradiationAnal cancerMost anal tumors arise fromThe standard treatment for anal canal tumors wasthe epidermal elements ofabdominoperineal resection, while anal marginthe anal canal lininggrowths were viewed as equivalent to skin tumors(squamous cell- 85% of analelsewhere and treated by local excision. Over thetumors), though some arisepast few years, radiotherapy and/or chemotherapyfrom the glandular mucosahave become increasingly popular and in manyof the uppermost part of thecases the treatment of choice.anal canal or from the analNon-surgical treatmentducts and glands(chemo-irradiation) for(adenocarcinomas).anal cancer has become increasingly popular. TheMalignant melanoma of thedrugs used are usually 5FU and mitomycin C. Theanus is very rare.radiotherapy consists of 30 Gy of external-beamTraditionally, the anal regionirradiation over a period of three weeks.is divided into the anal canalAlthought Adenocarcinoma is radiosensitive, it isand the anal margin or verge.usually treated by radical surgery.Esophageal cancerSquamous cell carcinomaSurgical resection is the treatment of choice foris the most commonearly stage disease. Advanced disease ishistological type, butchemosensitive Post-resection irradiationadenocarcinoma isimproves the loco-regional control in patientsincreasing in incidence..with positive resection margins, but not if there isnodal involvement. The dose is limited to 50 Gyin 25 daily fractions in patients who haveundergone a gastric pull-up or intestinalinterposition procedure.Pre-operative (neo-adjuvant) chemotherapy candownstage the primary tumor and treat micro-metastases before the post-operative, stimulatorysurge of growth factors. Drugs effective inesophageal cancer include: 5FU, cisplatin,mitomycin, paclitaxel, and methotrexate. Themost commonly used combination is continuousinfusion of 5FU and cisplatin.The majority of esophageal tumors are diagnosedat an advanced stage when resection is notpossible. Combined chemo-irradiation therapy ismore effective than radiotherapy alone in locallyadvanced esophageal cancer.Gastric cancerGlobally, stomach cancer isThe type of surgical resection used in gastricthe second commonestcancer will depend on the site of the tumor;cancer.esophogastrectomy may be required for cancers of90% of stomach tumorsthe esophagus/stomach/jejunum and proximalare adenocarcinomas,stomach; total gastrectomy for mid-stomachthat are futhertumors, and partial gastrectomy may be adequatedivided intotreatment for tumors in the distal stomach. A‘intestinal’ and ‘diffuse’futher consideration is the extent oftypes. The remainder arelymphandectomy undertaken.mainly lymphomas andRecent studies of post-operative adjuvantleiomyosarcomas. Intestinalradiotherapy are encouraging. Encouraging resultstype gastric cancers arehave also been seen with the use of mitomycin C,ulcerative and occur moretegafur, as well as ECF (epirubicin, cisplatin,often in the distal stomachprotracted venous infusion of 5FU) as post-than diffuse types, includingoperative adjuvant chemotherapy.linitinis plastica, whichoccur throughout thestomach.Small intestine andMore than 35 pathologicalSurgical resection is the most important therapycarcinoid tumorsvariants of small bowelfor all small bowel tumors. Chemotherapy fortumors have been described.most malignant small bowel tumors is basedMost common arearound 5FU applied either alone or inadenocarcinomas thatcombination with leucovorin and/or α-interferon.constitute 40% of allSarcomas are treated, as at other sites, withmalignant small boweldoxorubicin-based regimens. Similarly,tumors. Carcinoid tumorslymphomas are commonly treated with regimensrepresent 30% of all smallsuch as CHOP (cyclophosphamide, doxorubicin,bowel malignancies. Othervincristine and prednisone).forms of tumors areMost patients with low-grade carcinoid tumorslymphomas, sarcomas, ascan be treated with α-interferon alone or inwell as benign tumors suchcombination with somatostatin analogs.as leiomyomas, angiomasCytokine therapy (α-interferon) can be used bothand lipomas.for adenocarcinomas and carcinoids of the smallbowel, either alone or in combination with 5FU.Somatostatin analogs are useful for amelioratingclinical symptoms related to carcinoid tumors andalso demonstrate some anti-tumor effect.Cancer of the liverPrimary liver cancer orSurgery offers the only hope of cure for HCC.hepatocellular carcinomaHowever, resection is not possible in many(HCC) is prevalent in areaspatients. Liver transplantation is appropriate forwhere hepatitis B is endemic.some cirrhotic patients.Hepatitis C infection isFor irresectionable tumorsbecoming an increasingsystemic chemotherapy has been used.cause. Tumors may also beA variety of cytotoxic drugsfound incidentally at the timeincluding doxorubicin, mitozantrone,of post-mortem examinationmethotrexate and cisplatinum have been used asin patients with cirrhosis.single agents or in combination with 5FU.Surgical ligation of the hepatic artery andinsertion of a hepatic artery catherter have beenused to deprive the tumor of oxygenated bloodand to provide a route for the direct administrationof cytotoxic chemotherapy. Such techniques havebeen superceded by the use of chemo-embolization via radiologically placed catheters inthe hepatic artery. This technique is less invasiveand, using chemotherapy such as adriamycincombined with lipiodol uptake by the tumor, isdemonstratable on CT scan. Repeat embolizationis performed at 2-3 month intervals and tumorregression with associated regeneration of normalliver may render some tumors resectable.Cancer of the biliaryMicroscopically, tumors ofChonangiocarcinomas should be resected if theretractthe biliary tract areare no distant metastases and no irreparableadenocarcinomas of theinvolvement of the hepatic artery and portal vein.papillary, nodular, orIt is advisable to perform a full lymph-nodesclerosing type. Papillarydissection.tumors develop moreAlthough cholangiocarcinomas are not generallycommonly in the gall bladderconsidered chemosensitive or radiosensitive,or in the distal bile ductresponses to chemotherapy with cisplatin and 5FUsclerosing and nodularand folinic acid have been observed. Newer agentstumors, in the proximal bilesuch as gemcitabine and irinotecan are underduct. Adenocarcinomasinvestigation.without specific features areFor tumors of the gall bladder, incidental tumorsthe most common.diagnosed after a laparoscopic cholecystectomyTumors of the bile duct canneed no further action unless the gall bladder wasdevelop within the bead ofruptured. Tumors of more advanced stages shouldthe pancreas, in thebe treated with radical surgery. Post-operativehepatoduodenal ligament, orchemotherapy with cisplatin has yet to be provenat the level of the hepaticto provide beneficial results. Radiotherapy is usedhilum,. Hilarin cases of incomplete resection.cholangiocarcinomas(Klatskin's tumors) accountfor more than half in mostseries.Cholangiocarcinomas,especially of the sclerosingand nodular type, infiltratealong the walls of the ductsand the perineural tissuebefore obstructing the lumen.Metastases to the lymphnodes are seen in somepatients undergoing surgery.Direct duodenal invasion andperitoneal carcinomatisisoccur late.Tumors of the gall bladderinfiltrate the muscular wallof the gall bladder and theneighboring liver tissue, andspreads to the regionallymph nodes and to the liver.Distant metastases occurlate.Pancreatic cancerThe majority of exocrineResection is the only treatment that offers thetumors are ductalpossibility of a cure at this time. There is noadenocarcinomas and 1-2%standard chemotherapy for pancreatic cancer. Theare acinar; the remainder arenucleoside analogue, gemcitabine, may improveof diverse histology. Geneticsurvival compared to a simple 5FU regimen.abnormalities found inAdjuvant therapy using 5FU, doxorubicin, andpancreatic cancer include themitomycin C may also increase survival.K-ras oncogene (90-100%)Patients with good performance status andand mutations in p53 (60%),localized resectable tumor may be considered forp16 (80%), and SMAD4radiotherapy, with or without chemotherapy. 5FU(50%) tumor suppressorhas been shown to increase median survival aftergenes. Spread is mainly toradiotherapy (35-40 Gy). With 3D-CT planning ofretroperitoneal tissue, theexternal-beam radiotherapy it is possible toliver, and the peritoneum,deliver 50.4 Gy in 28 daily fractions to pancreaticwith distant metastases to thetumors without exceeding tolerance for thelung, liver, and the bone.adjacent stomach, bowel, or kidneys. Botheternal-beam radiotherapy (EBRT) and intra-operative radiotherapy (IORT) have been used inthe adjuvant settings.Thyroid cancersClassification of thyroidManagement of differentiated thyroid cancercancers is made on abegins with radical thyroidectomy. Radio iodinehistological basis:therapy (40-80 mCi = 1500-3000 MBq) to ablatedifferentiated (papillary orthe thyroid remnant follows the operation.follicular), anaplastic,External beam radiotherapy is delivered to thelymphoma, and medullary.neck where the tumor was locally invasive atLymphomas are high gradeoperation (giving a dose of 5000 Gy in 5 weeks).immunoblastic non-If the patient presents with metastatic disease, orHodgkin's tumors.at relapse after partial thyroidectomy, radicalMedullary carcinoma of thethyroidectomy and ablation is still required.thyroid arises from theFollowing radical thyroidectomy and radioiodineparafollicular C cells (theablation of the stump, the patient has at least twocell of origin of calcitonin).whole body radioiodine whole body profile scansFor recurrent disease, serumin the next 1½ years.calcitonin serves as a goodThe treatment of neck nodal disease is surgicalmarker,resection followed by radioiodine therapy whereappropriate. Metastatic disease further afield istreated by serial doses of radioiodine (150 mCi =5500 MBq). Iodine therapy is continued at 4-6monthly intervals until maximal remission. Inpatients who do not relapse, blood checks shouldbe made to ensure thyroid stimulating hormone(TSH) suppression and minimal serumthyroglobulin readings (<2 μg/L). For Anaplasticcarcinoma, radiotherapy to the neck is thestandard, with palliative chemotherapy as asystemic alternative. These tumors are rarely aradical surgical proposition and do not take upiodine.Lymphomasare treated with a combination ofcombination chemotherapy and wide fieldradiotherapy to the neck.For medullary carcinoma, surgical clearance ofapparently localized disease to the neck is the firsttherapeutic step. For recurrent disease,,occasional avidity of uptake tometaiodobenzylguanadine (MIBG) makesradioiodinated MIBG therapy a possibly usefultherapeutic modality.Adrenal cancerThe adrenal gland isThe mainstay of treatment is surgical resection.composed of a cortex and aFor phaeochromocytomas with residual ormedulla and the tumors thatunresectable disease, anti-hypertensive medicationarise in these two regions aremay be required to control the blood pressure. Ifaetiologically andthe tumor takes up the radionuclide MIBG, afunctionally different,therapeutic dose of up to 10,000 MBq of 131I-reflecting their cells ofMIBG may be administered. Although theseorigin. Adrenocorticaltumors were traditionally considered to be chemo-carcinomas are usuallyresistant, a combination of dicarbazine (DTIC),adenocarcinomas andvincristine, and cyclophosphamide shows activity.malignancy is confirmed byOther regimens which include cisplatin andthe presence of metastaticetoposide have shown promise.spread. These tumors extendFor unresectable or metastatic adrenocorticallocally to nodes and the livertumors, Mitotane (opDDD) is the first-linebut have a high frequency oftreatment. Metyrapone, antiglutethamide, anddistant spread. Adreno-ketoconazole are second-line medical therapies.medullary tumorsThe most active chemotherapy drugs appear to be(phaeochromocytomas) arecisplatin and etoposide. The role of externalrare, and can be difficult toradiotherapy is often for the treatment ofdiagnose.symptomatic metastases, usually in bone.NeuroblastomaNeuroblastoma is theSurguery is usual for tumors in the thorax. In thecommonest extracranialpast, later stage tumors were treated with intensivesolid tumor in childhood.chemotherapy. It is now recommended that, ifTumors arise in sympatheticpossible, surgical clearance be attempted, but ifnervous tissue. Morphologythis is not feasible then non-intensiveranges from a verychemotherapy, such as Vincristine, Cisplatin,undifferentiated, small,Etoposide and Cyclophosphamide (OPEC) orround cell tumor that may beVincristine, Carboplatin, Etoposide anddifficult to distinguish fromCyclophosphamide (OJEC), followed by delayedrhabdomyosarcoma to asurgery, be used.highly differentiatedMIBG posivity, post-operatively, indicates likelyganglioneuroblastoma.residual primary tumor, and local radiotherapy isindicated. An alternative is the use of I131 MIBG-targeted radiotherapy. High-dose melphalan withperipheral blood stem rescue is also used.Alternative high-dose intensity strategies, such asadministering treatment every 10 days orescalating the dose of cyclophosphamide are alsounder investigation.For late stages of disease occurring in infants lessthan 1 year of age, intervention withchemotherapy or radiotherapy may be appropriate.Low-dose irradiation to the liver or low-dosechemotherapy using vincristine or combinationsof cyclophosphamide, doxorubicin, orcarboplantin/etoposide are indicated.Renal cancerAdenocarcinomas make upRenal transitional cell carcinomas are managedthe vast majority of renalsimilarly to transition cell carcinomas of thecancers. They werebladder.previously known asResection of the tumor is the standard. In patients‘hypernephroma’ or ‘Grawitzwith limited metastatic disease, nephrectomy maytumors’. Transitional cellbe indicated. Adjuvant therapies consisting ofcarcinomas can arise withincytotoxic chemotherapies, endocrine therapy,the urothelium of the renalradiotherapy, and interferon have been tested inpelvis and represent thethis disease. Vinca alkaloids are the mostmajority of the remainingcommonly used chemotherapeutics. With respecttumors. Transitional cellto endocrine therapy, progestins are widely used,carcinomas arise in the renalalthough progestagens can also be useful.collecting system. RenalBiological therapy has been extensively tested intumors may invade locally orrenal cancer. Interleukin-2 (IL-2) is the mostmay metastasize to the lungs,widely tested biological agent, alone or inlymph nodes, bone andcombination with lymphokine-activated killerbrain.cells (LAK). Less toxic subcutaneous IL-2regimens are also effective and can be combinedwith interferon-α (IFN) and/or cytotoxics.Wilms' tumorWilms' tumor (WT) is anSurgical extirpation is the fundamental treatmentembryonal neoplasm arisingfor Wilms' tumor. Pre-operative chemotherapyin the kidneyClassicalmay be beneficial. The use of chemotherapy and‘triphasic’ WT has stromal,radiotherapy as adjuvants to surgery is an essentialblastemal, and epithelialpart of WT treatment. Chemotherapeutic agentselements. The bone-which have been employed include actinomycin Dmetastasizing renal tumor(AMD) plus vincristine (VCR), AMD plus VCR(clear cell sarcoma) andplus doxorubicin, and AMD plus VCR plusmalignant rhabdoid tumordoxorubicin plus cyclophosphamide.are pathologically andgenetically distinct entities.Cancer of the bladderThe majority of urothelialThe usual surgical procedure isand uretertumors are transitional cellcystoprostatectomy in male patients or anteriorcarcinomas. Pure squamousbladder exenteration in female patients, withcarcinomas anddissection of local lymph nodes. Radicaladenocarcinomas representradiotherapy based on CT scan has let toabout 5% of tumors,increasing precision and more accurate dosimetry.although metaplasia canUsually the entire bladder is treated to encompassoccur in a primarythe risk of sublinical disease at other sites in thetransitional tumor. There is aurothelium. The technique uses either three orlink betweenfour fields and the common fractionationandenocarcinoma of theregimens include treatment to a dose of 64 Gy inbladder and presentation of32 fractions over 6½ weeks or 55 Gy in 20the tumor in the bladderfractions over 4 weeks.dome, often associated withChemotherapy has been investigated as anpersisting urachal remnant.adjuvant therapy in localized disease either priorSquamous cancers of theto, or following, local treatment with surgery orbladder are associated withradiotherapy. Combination chemotherapy has anchronic bladder infection byestablished role in the palliation of patients withSchistosoma haematobium.incurable bladder cancer. Typical combinationsinclude M-VAC (methotrexate, vinblastine,adriamycin and cisplatin) and CMV (cisplatin,methotrexate and vinblastine). New drugs underinvestigation for the management of bladdercancer include taxanes, gemcitabine andifosphamide.Prostate cancerPathogenesis of cancer of theMeasurement of PSA should be done in allprostate gland is clearlypatients with outflow symptoms, and is routinelyandrogen-dependent. Sinceused as a screening tool in the US. PSA levels >4the development of assays tomg/ml are considered to be suspicious; transrectalmeasure prostate-specificultrasound and needle biopsy are called for inantigen (PSA) to diagnosethese patients. PSA levels >50 ng/ml may beprostatic disease, theindicative of distant metastases.condition has beenFor treatment of disease confined to the prostate,increasingly diagnosed at anradical local therapy (prostatectomy orearlier stage. More patientsradiotherapy) should be considered. Good resultsare presenting with earlyhave been seen with neo-adjuvant anti-androgenstage disease and suitabletherapy when followed by radiotherapy in locallytreatment strategies areextensive disease.required to cure those thatOnce the tumor has spread beyond the localcan be cured and palliatelymph nodes, useful palliation of the disease canoptimally those that cannot.be achieved by hormonal therapy. Treatmentsused to produce cessation of androgen-drivengrowth of the cancer include surgical castration,estrogens, steroidal anti-androgens such ascyproterone acetate, non-steroidal anti-androgens(flutamide, bicalutamide), and medical castrationwith LHRH agonists. The combination of medicalcastration and anti-androgen therapy has beenused.The chemotherapeutic anthracyclines and therelated mitoxantrone have been reported to beuseful. Radiation therapy offers useful palliationof advanced disease both in bone and soft tissue.In addition, radioactive strontium given by IVinjection (150 MBq) has proved effective inrelieving bone pain and delaying the progressionof symptomatic bone disease.Testicular cancerTesticular cancer is theThe management of seminoma and non-commonest cancer in youngseminomatous germ cell tumors depends on themen. The majority ofstage of the disease. Initial management oftesticular cancers are germtesticular germ cell tumors generally includescell tumors which arise ininguinal orchidectomy, although in cases withthe germinal epithelium.widespread metastases immediate chemotherapyBoth seminomas and non-may be appropriate. Carcinoma in situ willseminomatous germ cellprogress to invasive cancer, either seminoma ortumors are thought to arisenon-seminomatous germ cell tumors. Low-dosefrom pre-existing carcinomasratiotherapy to the testis (20 cGy in 10 fractionsin situ. Non-germ cellover 2 weeks) is useful in early stage patients.testicular tumors have beenStandard doses of radiotherapy (30 Gy in 15observed as well..fractions over 3 weeks) and chemotherapy areNon-seminomatous germ cellappropriate for later stage patients.tumors produce markers inthe form of the humanchorioric gonadatrophin(HCG) and/or alpha fetaprotein (AFP) in a majorityof cases. Seminomas have noreliable tumor marker withwhich to monitor disease,although HCG may be raisedin about 25% of cases. Thelactate dehydrogenase (LDH)may be raised in both tumorsas well.Ovarian cancerOvarian cancer is the fifthTreatment consists of surgery, followed for mostcommonest cancer inpatients with platinum-based chemotherapywomen. Over 80% ofregimens. For patients who cannot be optimallyovarian malignancies aredebulked at initial laparotomy, interval debulkingepithelial, the rest comprisesurgery after three cycles of chemotherapy confersgerm cell and stromala significant survival benefit. The standardtumors. Epithelial cancerschemotherapy regimen currently consists ofare classified by their cellcisplatin-paclitaxel, although carboplatin-type (serous, mucinous,paclitaxel has produced promising results. Aendometroid, Brenner,number of new agents, including topotecan,mixed) and by their grade.liposomal doxorubicin, etoposide, gemcitabine,80% of women withand altretamine also have shown response in thisadvanced ovarian cancersetting. A number of new approaches are alsohave elevated serum CA125under consideration, including intra-peritonealand this marker is valuablechemotherapy, high-dose systemic chemotherapy,in monitoring response tobiological response modifiers, metalloproteinasetherapy and for detection ofinhibitors, and anti-angiogenic agents.early relapse.Cancer of the uterineBy far the commonest cancerThe usual indication for radiotherapy incorpusof the uterine corpus isendometrial cancer is as adjuvant therapycarcinoma of thefollowing hysterectomy but less commonly it mayendometrium; less commonbe used as a primary treatment, as a follow-upare the uterine sarcomas. Oftreatment when residual tumor remains followingthe latter, some have asurgery, for treatment of inoperable tumors, orglandular element, the so-for treatment in cases of recurrence. Adjuvantcalled Mixed Mesodermalradiotherapy is generally delivered with external-Mullerian Tumors (MMMT)beam irradiation of a planned volume (usuallyor carcinosarcomas, andfour fields) to around 50 Gy over four weeks.some are fibrosarcomas.Chemotherapeutic combinations such as cisplatinThe epidemiology of uterineand doxorubicin have been shown to have somesarcomas is less well-definedactivity in endometrial cancer. Progestagens havethan endometrial carcinoma,been widely used in the past but should not bealthough cases of mixedused as long-term adjuvant therapy.Mullerian tumor have beenSuccessful treatment of MMTs depends onreported following tamoxifensurgery for localized disease. Residual disease ortherapy. The mixedtumors with nodal or distant metastases areMullerian tumors are dividedusually incurable. Hysterectomy and bilateralinto homologous andsalpingo-oophorectomy should be performed,heterologous. Homologoustogether with pelvic and para-aorticMMTs are composed oflymphandectomy. Residual disease can be treatedtissues native to the uterus,with radiation. Adjuvant radiation may be of use.whereas heterologous MMTsChemotherapy (e.g. doxorubicin and ifosfamide)involve tissue from outsideis often used in cases of metastatic disease.the uterus. Fibrosarcomasmay arise de novo or rarelyare found in a fibroid uterus.Cancer of the cervixIn developing nations,When the disease is confined to the cervix, patientcervical cancer is the mostmanagement depends on the cytology and/orcommon female cancer.histology specimens,Histology can reveal aThe type of surgical treatment to be used willspectrum of changes in thedepend upon the stage of the disease.epithelium of the cervix,Radiotherapy is typically used; a standard therapyincluding slight dysplasticconsists of external-beam irradiation (40-45 Gy tochanges in the cellthe pelvis for 4 weeks) followed by intra-cavityarchitecture, viralbrachytherapy (ICT) in the uterus/upper vagina orcytoplasmic changes, intra-to the central pelvic structures. Althoughepithelial neoplasia, micro-chemotherapy does not have an established role ininvasive carcinoma, andthe treatment of cervical cancer at this time,frank invasive carcinoma.patients with recurrent pelvic or systemicmetastatic disease may benefit from it. Theprincipal active agents are cisplatin, mitomycin C,ifosfamide, methotrexate, 5-fluorouracil, andbleomycin. Modern management of advanceddisease regularly involves the use of concurrent orsequential chemotherapy and radiotherapy.Vaginal and vulvarMost vaginal malignanciesRadical radiotherapy with a combination of pelviccancerare metastatic, from primaryexternal-beam and utero-vaginal intra-cavitarysites in the cervix, vulva,brachytherapy is the treatment of choice forendometrium, or trophoblastvaginal cancer. Lower vaginal involvement should(choriocarcinoma). The mostprompt consideration of either additional groincommon histological typesnode dissection or irradiation.of primary cancer arePrimary treatment of invasive vulvar cancersquamous andconsists of surgical excision with clear marginsadenocarcinoma.and removal of groin nodes. Chemo-irradiationIn primary invasive vulvar(typically 5FU and mitomycin C combined withcancer, the majority areradiotherapy) used in cases of advanced diseasesquamous carcinoma; othershows encouraging results.types include basalcarcinoma and malignantmelanoma.Trophoblastic tumorsGestational trophoblasticTreatment of hydatiform moles may includedisease (GTD) includes agentle suction curettage. Hysterectomy orspectrum of disorderscesarean section increases the risks ofranging from the pre-chemotherapy being required to eradicatemalignant completepersistent trophoblastic disease. The stage of thehydatidiform mole (CHM)disease is used to determine what chemotherapyand partial hydatidiformregimen is appropriate. Methotrexate is commonlymole (PHM), to theused in later stage patients. Combinationmalignant invasive mole,chemotherapy comprising etoposide,gestational choriocarcinoma,methotrexate, and actinomycin D (EMA)and the highly malignantalternating weekly with cyclophosphamide andplacental-site trophoblasticcincristine (CO) may be used in very late stagetumor (PSTT). Both CHMpatients. Treatment with methotrexate orand PHM can develop intoEMA/CO regimens continues until the HCG hasinvasive moles. However, itbeen normal for six weeks.is thought that only CHMChoriocarcinomaWbere it can be safely achieved,may progress to the highlyexcision biopsy of a metastasis should be used tomalignant choriocarcinomaconfirm the diagnosis. Patients are scored andand the rare PSTT.treated the same as for molar disease.Choriocarcinoma is a highlyThe behavior or PSTT is quite different frommalignant form thatother forms of GTD and it is relativelyusually presents within onechemoresistant. The best management isyear of pregnancy. One thirdhysterectomy when the disease is localized to theof patients present withuterus. When metastatic disease is present,metastases to the liver, brainpatients can respond to multi-agent chemotherapyor lung.either alone or in combination with surgery.PSTT can develop followinga term delivery, non-molarabortion, or CHM. PSTTsare slow-growing malignanttumors composed mainly ofcytotrophoblast with verylittle syncytiotrophoblast, soproducing little HCG.However, they often stainstrongly for human placentallactogen (HPL) which helpsdistinguish this tumor fromcarcinomas, sarcomas,exaggerated placental-sitereaction, and placentalnodule. In most cases spreadoccurs by local infiltrationwith distant metastasesoccurring late via thelymphatics and blood.Cancer of the larynxTumors arising from theThe main treatment modalities are radiotherapymucosa of the larynx makeand surgery. Induction chemotherapy may selectup the majority of tumors inpatients with advanced carcinoma suitable forthe larynx.full-course radiotherapy to save the larynx. Thevoice may be better spared by radiotherapy thansurgery.Cancer of the oralSquamous cancers of the oralSurgery is the most common treatment for earlycavitycavity are associated withtumors; primary radiotherapy and reserve surgeryheavy consumption of bothfor later stage disease. Locally advanced disease istobacco and alcohol. Thebest treated by a combination of surgery andother less commonradiotherapy. Doses of 50-65 Gy are delivered tomalignancies of the oralthe tumor volume, depending .on the extend of thecavity include salivaryresidual tumor. At least part of this dose can betumors (adenoid cystic,delivered by interstitial therapy. As with head andmucoepidermoid, andneck squamous cancers at other sites,adenocarcinoma),chemotherapy (cisplatin, 5FU, methotrexate,melanomas, plasmacytomas,bleomycin) is active in advanced disease.and sarcomas.Carcinoma of theNPC spreads into theNPC is generally both radio- and chemosensitive.nasopharynxparapharyngeal spaceRadiotherapy is the mainstay of treatment. Thelaterally. Any of the cranialrole of surgery is restricted to staging and thenerves can be involved in theelective dissection of neck nodes. Radiotherapytumor. Cervical lymph nodeplanning is complicated in this disease due to itsspread is common. Distantlocation in the body. Brachytherapy may be usedspread to bone, the liver andto increase the dose to the primary site. Local orlung is common in advancedregional recurrence can be treated by a secondstage disease,radical course of radiotherapy. There may beadvantages to using chemotherapy prior toradiotherapy in this disease. Commonly usedchemotherapy agents include 5FU, cisplatin andbleomycin. Combined radiation and chemotherapyregimes show promise.Cancer of the nasalThe sinonasal region offers aSurgery alone or in combination with radiotherapycavity and paranasalgreat histological diversity ofis required in the majority of cases. Radiotherapysinusestumors, with squamous cellmay be given before or after surgery wherecarcinoma the most common.appropriate. Megavoltage photons are optimallyDue to the intimateused, in a dose of 60-66 Gy in 30-33 fractionsrelationship between theover 6-6.5 weeks. Concurrent chemo-irradiationnasal cavity and sinuses andtherapy is under investigation. A number ofthe orbit and skull base, thechemotherapeutic agents (cisplatin, 5FU,tumors may spread early tomethotrexate, bleomycin) have significant activitythese areas. Additionally,in recurrent or metastatic disease.anterior spread into the softtissues of the face, posteriorinvolvement of the pterygoidregion and nasopharynx, andinferior spread to the oralcavity are also commonlyencountered. Distantlymphatic and hematogenousspread are rare in the earlystages of disease.Basal cell carcinomaBasal cell carcinomas occurRadiotherapy can be used as primary treatment,of the external eye-frequently on the face andfollowing excision when the surgical margins areskin, lids, lacrimalparticularly around the eye.positive, or where a recurrence has occurred.glandsThey have a prediliction forSuperficial X-rays (100-150 kV) are appropriatethe lower lid margin and(e.g. 45 Gy in 10 daily fractions).adjacent inner canthus. It isunusual for the upper eyelidto be involved. Surgicalexcision is often appropriate.In more extensive lesions orwhere surgery iscontraindicated, radiotherapycan be given.Non-Hodgkin'sLymphoma may involve theLow-grade lymphoma is treated with localLymphoma of the eyeupper and lowerradiotherapy; high-grade lymphoma with localizedand orbitlids/conjunctiva, majordisease is treated with chemotherapy followed bylacrimal gland, or orbit.local radiotherapy. Other stages are treated withchemotherapy.Squamous andSquamous carcinoma arising from the skin, lids,adenocarcinoma of theor very rarely the conjunctiva can be treated witheye and orbitradiotherapy. Sebaceous and lacrimal glandtumors should be treated primarily with surgery.Melanoma of the intra-Melanoma can affect theTreatment may be observation, radioactive eyeocular regionuveal tract; its most frequentplaque (ruthenium or iodine), local resection,location is the choroids.charged particles (proton beam), or enucleation.Local resection and proton-beam irradiation areused for larger tumors.RetinoblastomaThis is a rare intra-ocularPhotocoagulation can be used to treat smalltumor arising in youngtumors not adjacent to the macula or optic disk.children, usually in the firstSmall to moderate tumors can be treated withtwo years of life,radioactive plaques. External radiotherapy is usedto treat large or multiple tumors. It may benecessary to radiate the entire eye (40 Gy in 20fractions over 4 weeks). These tumors are oftenchemosensitive, and chemotherapy can beparticularly useful as a neo-adjuvant or when thetumor has a poor prognosis. Usefulchemotherapeutic agents include platinum,etoposide, vincristine, doxorubicin andcyclophosphamide.Optic nerve tumorsOptic nerve tumors are rare and are usually treatedby surgery. When excision is not possible, high-dose radiotherapy can be given, usually usingpaired wedge fields and giving doses on the orderof 55 Gy conventionally fractionated.Rhabdomysarcoma ofThis rare tumor is mostthe intra-ocular regionfrequently located in theorbit.Metastatic disease ofMetastatic disease involvingTreatment with radiotherapy should usually bethe intra-ocular regionthe eye is usually associatedwith a lateral field to the orbit giving 20 Gy in fivewith choroidal metastases.fractions over a week. The field may be angledThe commonest tumorsfive degrees posteriorly to avoid the contralateralimplicated are lung andlens.breast.Widespread bone metastases may affect the eye.Systemic treatment is often appropriate, althoughlocal radiotherapy may be helpful.Salivary gland tumorsThe most common tumor ofIn treatment of malignant salivary gland tumors,the parotid gland is theadjuvant radiotherapy is most commonlypleomorphic adenomal, alsodelivered post-operatively, but also as a split-known as the mixed parotidcourse, giving two-thirds of the treatment beforetumor. Ocasionally thisand one-third of the treatment after the operation.benign tumor can undergoPost-operative radiotherapy reduces the incidencemalignant transformation.of local recurrence. Orthodox radiotherapyMost acinic cell andtechniques use 6-10 MV photons, often with amucoepidermoid tumors arewedged pair of fields, to give a dose of 55-65 Gylow grade, although someover 6-7 weeks. More aggressive tumors includingmay be high grade and moremucopidermoid, adenoid cystic, andaggressive with localundifferentiated carcinomas appear initially toinvasion and a tendency torespond well to chemotherapeutic agents. Themetastasize to lymph nodesmost commonly used agents are cisplatin andand through the bloodstream.5FU.Wide removal isrecommended forpleomorphic adenomas andacinic cell andmucoepidermoid tumors, andwill cure the majority oftumors. Carcinomas exhibitmore aggressive behavior.Primary brain tumorsPrimary brain tumorSurgery and radiotherapy remain the mainstays ofpathology is extremelytreatment for these tumors and most advancesvaried reflecting diverseinvolve applying these modalities more effectivelyhistogenesis. Gliomas are theto more precisely delineated tumors. Post-most common tumors.operative radiotherapy is normally given. The areaPrimary brain tumors rarelyirradiated us usually limited to tumor-bearingmetastasize outside the CNSbrain and doses of 30-60 Gy are given over 2-6but are highly infiltrative,weeks. Chemotherapy is usually reserved forwith a tendency to spreadrelapse. The nitroureas including carmustinealong white matter tracks to(BCNU) and lomustine (CCNU) are the mostmore distant regions of thecommon agents used. Procarbazine and thebrain. Spread throughplatinum compounds and temozolomide may alsocerebrospinal fluid (CSF) tohave some value. Following surgical clearance theremote areas of the neuraxisapplication into the resection cavity of sustained-is a feature of germ cellrelease chemotherapy (BCNU-Gliadel) has showntumors, medulloblastoma,some benefit. Third generation radiationand other primitivesensitizers such as the hypoxic cell cytotoxinneuroectodermal tumors.tirapazamine have shown promise. Angiogenesisinhibitors may be of use, and the drug thalidomidehas shown some activity. Modificationof the localenvironment with O-6-alkylguanine DNAalkyltransferase inhibitors or blood-brain barriermodifiers may improve results withchemotherapeutic agents.Brain metastasesMetastases to the brain fromInitial management requires control of thean extracranial primary sitepresenting symptoms with medication.is common.Dexamethasone is indicated in the majority ofpatients. A starting dose of 8-16 mg per daily iscommon and improvement can often bemaintained with doses of 2-4 mg. For patientswhose condition is improved following treatmentwith dexamethasone, radiotherapy can be offered.Typically, the whole brain is irradiated, with adose of 20 Gy in five fractions being effective.When a solitary metastasis is present, tumorresection, if possible, is the treatment of choice.Post-operatively, whole-brain radiotherapy isgiven (20 Gy in 5 fractions or 30 Gy in 10fractions) with or without a local boost.Chemotherapy can be useful in patients with brainmetastases from chemo-sensitive primary tumors.It should be used as a first-line therapy (orfollowing resection) in germ cell tumors or as analternative to radiotherapy in small cell lungcancer or lymphoma. It can also be used assecond-line treatment in less chemo-sensitivetumors such as breast cancer.Primary cutaneousPrimary cutaneous malignantTreatment is complete excision of the lesion. Latemalignant melanomamelanoma arises fromstage melanoma may require dissection ofmelanocytes in the basal skinregional lymph nodes. Adjuvant alpha interferonlayermay be of use. Chemotherapeutic regimens includeDTIC and/or vindesine; the addition of tamoxifenmay be of use. Interleukin 2 combined with withmelanoma-directe vaccines are being investigated.Radiotherapy is generally reserved for palliationin metastasized cancer.Non-melanoma skinCommonest cancer inStandard treatment of basal cell carcinomas iscancersWestern populations.surgical excision. Metastases are rare and areBasal cell carcinomas ariseusually curable by surgical excision oron sun-exposed areas.radiotherapy. Other treatment strategies includeVariant lesions includecryosurgery and topical 5FU.nodular, ulcerative,Treatment of squamous cell carcinoma is eitherpigmented, superficial,surgery or radiotherapy. Chemotherapy has beencystic, morphoeic, andused for disseminated disease (cisplatin,multicentric.methotrexate, 5FU, bleomycin).Squamous cell carcinomaTreatment of Merkel cell carcinoma is surgical.also arises on sun-exposedPalliative radiotherapy is useful in metastaticsites but is faster growing.disease.May metastasize to regionalnodesMerkel cell carcinoma is arare but highly malignantneuroendocrine tumor,commonly spreads toadjacent skin and regionalnodes.Apocrine and eccrine glandcancers are rare and usuallyonly locally invasive.Other uncommon skinmalignancies includecutaneous angiosarcoma,Kaposi's sarcoma (HIV-associated or endemic), andother soft tissuemalignancies.Acute lymphoblasticALL is the commonestTreatment of ALL includes:leukemia (ALL)cancer in children.Chemotherapy: induction of remission is routinelyachieved by combining vincristine, prednisolone,and 1-asparginase. Additionaly anthracycline isused in adults. Consolidation is a crucial phasefollowing remission. Exposure to new drugs is akey strategy (cyclophosphamide, thioguanine,cytosine arabinoside). Clearance of the centralnervous system (CNS) as a sanctuary site is alsokey. CNS irradiation or MIX intra-thecally orhigh-dose IV chemotherapy are used.Maintenance of patients in remission is achievedusing a cyclic schedule of methotrexate, 6thioguanine, vincristine and prednisolone.Treatment of high-risk disease involvesintensification in consolidation withcyclophosphamide or methotrexate in higherdoses along with stem cell transplantation in firstremission. Treatment following long remissionconsists of further chemotherapy or stem celltransplant.Acute mycloidAntracycline and cytosine arabinose given overleukemia (AML)7-10 days is the backbone of treatment. Addition ofa third drug (thioguanine or etoposide) is widelyused. Giving higher ara-C doses in induction maybe of benefit. Following induction, three or fourfurther intensive courses incorporating other drugs(e.g. amasacrine, etoposide, idarubicin,mitoxantrone, and araC at higher doses) areusually given. Maintenance therapy no longerstandard but may be useful, along with stem celltransplantation.Acute promyelocyticAll-transretinoic acid (ATRA) used alone canleukemiainduce remission. Additional chemotherapy, givenalone or subsequent to ATRA is essential.Autologous transplantation or allogenic BMTwidely used to consolidate first remissions.Arsenic compounds may be of use, as may refinedstem cell transplantation and immunologicallybased approaches.Chronic lymphoidChronic lymphoid leukemiasTreatment of B-cell chronic lymphocytic leukemialeukemiasare a heterogenous group ofmay not prolong survival of patients withconditions associated withlymphocytosis or uncomplicatedaccumulation of lymphoidlymphandenopathy. Systemic therapy is indicatedcells in the peripheral bloodfor advanced disease or in cases with diffusethat may infiltrate into theinfiltration or low lymphocye doubling time.bone marrow.Chlorambucil is first-line therapy. Single-agentB-cell chronicprednisolone (1 mg/kg/day) is useful in advancedlymphocyticdisease. Combination chemotherapy using COP orleukemia is the commonCHOP has been evaluated.. Purines areleukemia of late middle-age.considered the best second-line therapy. Singleagent fludarabine standard. Chlorambucil alsoused at a dose of 25 mg/m2 IV for 5 days on a 4-6week cycle for up to 6 cycles. Radiotherapy iseffective local treatment for lymph nodes.Splenectomy is effective for massivesplenomegaly, anemia or thrombocytopenia andfor conditions refactory to prednisolone andcytotoxic therapy.Hodgkin's diseaseStandard therapy for early stage Hodgkin'sdisease comprises nodal irradiation.Chemotherapy combined with limited (involvedfield) radiotherapy has been investigated.Chemotherapy regimens include VBM(vinblastine, bleomycin, methotrexate) and ABVD(doxorubicin, bleomycin, vinblastine,dacarbazine). In cases with good prognosisinvolved field radiotherapy has been used alonesuccessfully.For Advanced stage Hodgkin's disease, MOPP(mustine, vincristine, procarbazine, prednisolone)and its variants were standard until recently.Doxorubicin-based chemotherapy (particularlyABVD) is now considered standard. Newlyintroduced brief-duration regimens include:Stanford V (mustine, doxorubicin, vinbiastine,prednisolone, vincristine, bleomycin, etoposide);BEACOPP (bleomycin, etoposide, doxorubicin,cyclophosphamide, vincristine, procarbazine,prednisolone); and may be combined with limitedfield radiotherapy to sites of bulk disease.For salvage therapy in Hodgkin's disease, relapseafter radiotherapy frequently responds tochemotherapy. Relapse after first linechemotherapy frequently responds to second linechemotherapy. High-dose chemotherapy andautologous stem cell transplantation becomingstandard.Other promising therapies for Hodgkin's diseaseinclude high-dose chemotherapy and antibodiesagainst CD30 antigen on R-S cell.Non-Hodgkin'sNon-Hodgkin'sFor low grade NHL, treatment is reserved untillymphomas(NHL)lymphomas(NHL) are aactive symptoms present. Local radiotherapy isgroup of malignant diseasesapplied to bulky sites. Single-agent chemotherapyarising from cells of the(chlorambucil), as well as combination therapyimmune system.(CHOP) are standard. New treatments includeHigh grade NHL ispurine analogs (fludarabine, 2-CDA), autologousconsidered to have a strongtranplant, alpha interferon, palliation with anti-tendency to involve the CNSCD20 antibody (rituximab), antisense therapy toand is comprised of Burkitt'ssuppress Bcl-2 protein.lymphoma, lymphoblasticBurkitt's lymphoma is treated with intensivelymphoma, and diffuse largechemotherapy with methotrexate,B-cell NHL. Diffuse large B-cyclophosphamide, ifosamide, and withcell NHL is the commonestintrathecal therapy.high-grade NHL.Lymphoblastic lymphoma treatment includesemergency management of mediastinalobstruction and prevention of tumor lysissyndrome. Intensive combination chemotherapyschedules similar to ALL including CNS-directedtherapy have been used. Allogenic and autologousprogenitor cell transplantation may improvesurvival.Radiotherapy can cure early stages of diffuselarge B-cell NHL. CHOP chemotherapy withradiotherapy to bulk disease is standard. Relapsedpatients may respond to salvage chemotherapyand may benefit from autologous progenitor celltransplantation.MyelomaMyeloma (multipleChemotherapy (oral mephalan, 4-day pulses everymyeloma, myelomatosis) is4 weeks for 3 months) is usually used fordue to unregulatedpalliation. More intensive chemotherapy (VAD,proliferation of monoclonalvincristine and adriamycin by IV infusion an oralplasma cells in bone marrow.dexamethasone) may produce response.Combination chemotherapy (e.g. doxorubicin,BCNU, cyclophosamide, melphalan) may produceresponse as well. High-dose (marrow ablative)melphalan with autologous stem cell transpant canbe considered in younger patients. Allogenictransplantation has been reported to improvesurvival. Oral clodronate may reduce the bonefracture rate when dosed continuously, as maypamidronate. Interferon-a may be used asmaintenance therapy. l3rythropoietin may be usedto reduce transfusion requirementsOsteosarcomaOsteosarcoma is theSurgical resection and use of chemotherapy in thecommonest primary boneadjuvant (post-operative) or neo-adjuvant (pre-tumorand post-operative) setting has become standard.The most active chemotherapeutics includedoxorubicin, cisplatin, ifosamide, and high-dosemethotrexate. Osteosarcoma is relatively radio-resistant: radiotherapy rarely used as primarytreatment, limited to palliative treatment or inmetastases, when combined chemo-irradiationtherapy is useful.Ewing's sarcomaChemotherapeutic agents with substantial activityin Ewing's sarcoma include vincristine,doxorubicin, actinomycin D, etoposide,cyclophosphamide, and ifosfamide. Surgeryand/or radiotherapy are used to treat the primarytumor. Lung metastases should be treated withconventional chemotherapy with whole-lungirradiation to any residual disease. Bone metasasesmay benefit from megatherapy after conventionalinduction treatment-Melphalan, busulphan, TBI,or a combination may be-used as a conditioningregimenPrimary malignantPrimary malignant spindleTreatment is surgical removal. Chemotherapy isspindle cell sarcoma ofcell sarcoma of bonebeing explored and may be of benefit.boneincludes liposarcoma,angiosarcoma,leiomyosarcoma, andhemangiopericytoma.OsteoclastomaOsteoclastoma is usuallyTreated by curettage and bone packing or bonebenign.resection and insertion of an endoprosthesis.Radiotherapy risks malignant transformation.Response to chemotherapy has been reported.ChondrosarcomaFor low grade chondrosarcoma tumors, treatmentis surgical resection with radiotherapy. For highgrade tumors treatment is combined surgery,chemotherapy and radiotherapy.ChordomaSurgery is treatment of choice. Radiotherapy(55-60 Gy) used after incomplete resection or aspalliation. Particle therapy with protons hasshown promise.Solitary plamacytomaTreatment is surgical resection or radiotherapy(45-50 Gy).Primary boneLocalized high-grade treated with initiallymphomachemotherapy (CHOP) followed by eitherradiotherapy or surgical excision withendoprosthesis. Multifocal or low-grade diseasetreated with chlorambucil chemotherapy andradiotherapy.RhabdomyosarcomaRhabdomyosarcoma (RMS)Treatment usually begins with combination(RMS) of the softof the soft tissue ischemotherapy followed by local treatmenttissuecomposed of embryonal(surgical resection with radiotherapy if excision isRMS and alveolar RMS.incomplete; radiotherapy alone if unresectable).Embryonal RMS may be treated with vincristineplus actinomycin D; may intensify treatment withifosfamide, doxorubicin, other drugs. Additionaldrug such as etoposide and carboplatin and high-dose alkylating agent therapy may be of use.Highly sensitive to radiotherapy; common doseswith chemotherapy are 40-50 Gy.Soft tissue Ewing'sMuch of management of RMS applies tosarcoma and primitivemanagement of the Ewing's family of tumors. Pre-neuroectodermal tumorand post-operative combination chemotherapy is(PNET)crucial (doxorubicin, actinomycin D, ifosfamide,vincristine). Surgery is the preferred localtreatment but radiotherapy is required afterincomplete resection or for unresectable tumors.Adult soft tissueRadical surgery is standard for the primary softsarcomastissue sarcomas (STS). Radiotherapy may reducelocal recurrence. Metastases may be surgicallyremoved from the lungs. Chemotherapy for locallyrecurrent or metastatic disease is largely palliative.Ifosfamide and doxorubicin are the most activechemotherapeutic agents. Combinationchemotherapy may be of use. Synovial sarcoma ischemosensitive. Adjuvant chemotherapy is notstandard but may be useful.Cancer of unknownCancer of unknown primaryEffective palliation with chemotherapy isprimary sitesite is categorized in fourindicated if the tumor is chemo-responsive.broad groups:Combination chemotherapy has been investigated.adenocarcinoma, poorlyHormone therapy is valuable in breast, prostate,differentiated carcinoma,and endometrial cancer. Thyroidectomy andundifferentiated malignancy,radioactive iodine should be used in thyroidand squamous carcinoma.cancer. Radiotherapy can be palliative in bonemetastases, brain metastases and for lymph nodemasses.ParaneoplasticParaneoplastic syndromessyndromesare pathological conditionscaused by a cancer but notdue to direct local infiltrationor metastatic spread. Cancerscommonly associated withparaneoplastic syndromesinclude: lung (SCLC andNSCLC), pancreatic,lymphoma (NHL and HD),breast, prostate, and ovary.EndocrineEndocrine paraneoplasticEndocrine paraneoplastic syndrome is treatedparaneoplasticsyndrome is a syndrome ofwith fluid restriction and democycline (150-syndromeinappropriate anti-diuretic300 mg 8 hourly).hormone (arginine-Specific antibody treatments are used to treatvasopressin). Other relatedCushing's syndrome. Standard treatment is toparaneoplastic syndromesdecrease cortisol secretion surgically or medicallyinclude Cushing's syndrome,(metyrapone, ocreotide, ketoconazole).hypercalcemia,Hypercalcemia is treated with saline hydrationhypocalcemia, andand IV pamidronate (60-120 mg).hypoglycemia.Hypocalcemia is treated with calcium infusions.Cushing's syndromeHypoglycemia is treated with glucose infusionsinvolves inappropriateand tumor debulking.overproduction ofadenocorticotrophichormone precursors.Hypercalcemia is due totumor production ofparathyroid hormone-relatedprotein.Hypocalcemia is associatedwith bone tumors andcalcitonin-secretingmedullary carcinomas of thethyroid.In hypoglycemia, the mostlikely cause is tumorproduction of the precursorto insulin-like growth factorII.NeurologicalNeurological paraneoplasticPeripheral neuropathy is treated withparaneoplasticsyndromes includecorticosteroids along with treatment for thesyndromeperipheral neuropathy,underlying cancer.encephalomyelopathy,Encephalomyelopathy is treated with anti-tumorparaneoplastic cerebel lartherapy.degeneration, cancer-Paraneoplastic cerebellar degeneration isassociated retinopathy,responsive to anti-tumor treatments, steroids, andLambert-Easton myasthenicplasmapherisis.syndrome, andCancer-associated retinopathy is treated withdermatomyositis/corticosteroids.polymyositis.Peripheral neuropathy isLambert-Easton myasthenic syndrome is treatedcaused by axonalwith standard cancer treatment, corticosteroids,degeneration orand plasma exchange.demyelination.Dermatomyositis/polymyositis is treated bysearching for and treating the tumor along withcorticosteroids, and/or azathioprine.HematologicalHematologicalErythrocytosis is treated with phlebotomy.paraneoplasticparaneoplastic syndromesHemolytic anemia is treated by treating the tumor;syndromesinclude red cell disordersreplacing coagulation factors, and supplying IV(erythrocytosis, hemolyticheparin.anemia, red cell aplasia) andwhite cell disorders(autoimmune neutropenia,platelet disorders,coagulopathy). Hemolyticanemia can be autoimmuneor micro-angiopathic.DermatologicalDermatologicalparaneoplasticparaneoplastic syndromessyndromesinclude pruritic conditions;pigmentation conditions(Acanthosis nigricans,Vitiligo); erythematousconditions (necrolyticmigratory erythema,exfoliative dermatitis-cutaneous); and bullousconditions (pemphigus,dermatitis herpetiformis).Other paraneoplasticOther paraneoplasticTreatment modalities for hypertrophicsyndromessyndromes includeosteroarthropathy include anti-tumor therapy,hypertrophicnon-steroidal antiinflammatories, corticosteroids,osteroarthropathy.and radiation.AIDS-relatedAIDS-related malignanciesThe primary goal of treatment of Karposi'smalignanciesinclude Karposi's sarcoma,sarcoma is to prolong life. Localized treatmentsnon-Hodgkin's lymphoma,include cryotherapy and laser, intra-lesionalprimary central nervouschemotherapy with vinblastine or interferon, andsystem (CNS) lymphoma,radiotherapy. Systemic treatment is required ifHodgkin's disease, cervicalthere is widespread cutaneous or visceralcancer, and anal cancer.involvement. Immunotherapy with interferon iseffective. Treatment with vincristine andbleomycin is also used; doxorubicin may beadded. Liposomal chemotherapeutics lookpromising. Treatments such as taxol, beta HCG,trans-retinoic acid, and anti-angiogenic agentssuch as thalidomide are being tested.For treatment of non-Hodgkin's lymphoma,systemic treatment with chemotherapy is usuallyused. Radiotherapy may be useful in localizeddisease: Standard treatment for advanced diseaseis combination chemotherapy, typically CHOP.Comcomitant intrathecal chemotherapy withmexotrexate and cytarabine is also used. Moreaggressive chemotherapy regimens may be useful.For primary CNS lymphoma, treatment involveswhole-brain radiotherapy in combination withsteroids and perhaps intrathecal chemotherapy.Hodgkin's disease is treated by standardchemotherapy is with ABVD (adriamycin,bleomycin, vinbiastine, dacarbazine).


[0152] The formulations of the present invention comprise a therapeutically effective amount of the nitric oxide mimetic formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term “pharmaceutically acceptable carrier” means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate. Coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the formulation according to the judgment of the formulator. The formulations of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), supralingually (on the tongue) sublingually (under the tongue), bucally (held in the buccal pouch), or as an oral or nasal spray. The oral spray may be in the form of a powder or mist which is delivered to the deep lungs by oral inhalation.


[0153] In yet other embodiments, the invention provides topical sustained and prolonged release pharmaceutical compositions comprising one or more pharmacological compounds described supra, and a pharmaceutically acceptable carrier, to treat a malignant cell phenotype disorders. One example of a topical formulation includes 75% (w/w) white petrolatum USP, 4% (w/w) paraffin wax USP/NF, lanolin 14% (w/w), 2% sorbitan sesquioleate NF, 4% propylene glycol USP, and 1% compound of the present invention. Such compositions are useful in the treatment of cancer and in controlling and reducing pain associated therewith. Such compositions may comprise a unit dosage of one or more particular active agent(s) (e.g., a NO donor, calcium channel blocker, cholinergic modulator, α-adrenergic receptor antagonist, β-adrenergic receptor agonist, phosphodiesterase inhibitor, cAMP-depedent protein kinase activator, superoxide scavenger, potassium channel activator, SOC inhibitors, benzodiazepine, adrenergic nerve inhibitor, antidiarrheal agent, HMG-CoA reductase inhibitor, smooth muscle relaxant, adenosine receptor modulator, adenylyl cyclase activator, cAMP mimetic, endothelin receptor antagonist, bisphosphonate, cGMP-dependent protein kinase activator, and cGMP mimetic. Preferably, the compositions are administered in unit dosage form to a subject in need of such treatment. Topical sustained and prolonged release compositions are typically variants which include 1) an absorbent in a hydrophilic base; 2) an absorbent in a hydrophobic base; and 3) coated beads containing an absorbent matrix dispersed in a suitable vehicle. Also provided are methods of treating cancer comprising topically administering an effective amount of such compositions (e.g., in unit dosage form) to the appropriate area of the subject in need of such treatment.


[0154] Such hydrophilic compositions and preparations of the invention comprise a compound of the invention and a polymer, such as cellulose (methyl cellulose, ethyl cellulose, hydroxy propyl cellulose, etc.), higher molecular weight polyethylene glycol, methacrylic-acrylic acid emulsion, hydrogel, carbopol, ethyl vinyl acetate copolymer, or polyester, etc., to bind the compound of interest to the polymer. The compound-polymer matrix is then dispersed in a hydrophilic vehicle to form a semi-solid. After administration of such hydrophilic composition into the appropriate area, the water in the semi-solid preparation is adsorbed and the polymer matrix with the active ingredient (i.e., the pharmaceutical compound) remains as a coating in the area to which it has been applied. The pharmaceutical compound is then slowly released from this coating.


[0155] Hydrophobic compositions and preparations of the invention employ similar polymers as used in the hydrophilic preparations, but the polymer/compound matrix is dispersed into a vehicle, such a plastibase, in the hydrophobic compositions and preparations. Plastibase is a mineral oil base that only partially dissolves the pharmaceutical compound. The semi-solid composition forms a thin coating to which the composition has been applied such as, e.g., the vagina or urethral tract) and slowly releases the active compound. The prolonged action is controlled principally by the solubility of the active ingredient in the vehicle.


[0156] The present invention also provides coated beads which are produced by first absorbing a compound of the present invention, or a combination of compounds, on a cellulosic material blended with polyethylene glycol, filler, binder and other excipients. The resulting matrix is then extruded and spheronized (e.g., the process of making into spheres) to create small beads. The beads are then coated to an appropriate thickness with one or more of a suitable material, such as a methacrylic-acrylic polymer, polyurethane, ethyl vinyl acetate copolymer, polyester, silastic, etc. The coating on the beads acts as a rate controlling membrane that regulates the release of the compound from the core beads.


[0157] In other embodiments, the invention provides pharmaceutical compositions suitable for oral administration which are provided in unit dosage form comprising per unit dosage a NO donor, calcium channel blocker, cholinergic modulator, α-adrenergic receptor antagonist, β-adrenergic receptor agonist, phosphodiesterase inhibitor, cAMP-dependent protein kinase activator, superoxide scavenger, potassium channel activator, SOC inhibitors, benzodiazepine, adrenergic nerve inhibitor, antidiarrheal agent, HMG-CoA reductase inhibitor, smooth muscle relaxant, adenosine receptor modulator, adenylyl cyclase activator, cAMP mimetic, endothelin receptor antagonist, bisphosphonate, cGMP-dependent protein kinase activator, cGMP mimetic, and a pharmaceutically acceptable carrier. Such compositions are useful for treating cancer, including those disorders and conditions described above.


[0158] For delivery to the buccal membranes, typically an oral formulation, such as a lozenge, tablet, or capsule is used. Methods of manufacture of these formulations are known in the art, including but not limited to, the additionof a pharmacological agent to a pre-manufactured tablet; cold compression of an inert filler, a binder, and either a pharmacological compound or a substance containing the compound (as described in U.S. Pat. No. 4,806,356); and encapsulation. Another oral formulation is one that can be applied with an adhesive, such as the cellulose derivative, hydroxypropyl cellulose, to the oral mucosa, for example as described in U.S. Pat. No. 4,940,587. This buccal adhesive formulation, when applied to the buccal mucosa, allows for controlled release of the pharmacological agent into the mouth and through the buccal mucosa. The compounds of the present invention can be incorporated into these formulations as well.


[0159] For delivery to the nasal or bronchial membranes, typically an aerosol formulation is employed. The term “aerosol” includes any gas-borne suspended phase of the pharmacological agent that is capable of being inhaled into the bronchioles or nasal passages. Specifically, aerosol includes a gas-borne suspension of droplets of the compounds of the instant invention, as may be produced in a metered dose inhaler or nebulizer, or in a mist sprayer. Aerosol also includes a dry powder composition of a pharmacological compound of interest suspended in air or other carrier gas, which may be delivered by insufflation from an inhaler device, for example. For solutions used in making aerosols, the preferred range of concentration of the pharmacological agent is 0.1-100 mg/ml, more preferably 0.1-30 mg/ml, and most preferably, 1-10 mg/ml. Usually the solutions are buffered with a physiologically compatible buffer such as phosphate or bicarbonate. The usual pH range is 5 to 9, preferably 6.5 to 7.8, and more preferably 7.0 to 7.6. Typically, sodium chloride is added to adjust the osmolarity to the physiological range, preferably within 10% of isotonic. Formulation of such solutions for creating aerosol inhalants is discussed in Remington, Pharmaceutical Sciences, see also, Ganderton, et al., Drug Delivery to the Respiratory Tract, Ellis Horwood (1987); Gonda, Critical Reviews in Therapeutic Drug Carrier Systems, 6:273-313 (1990); and Raeburn, et al., J. Pharmacol. Toxicol. Methods, 27:143-159 (1992).


[0160] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral formulations can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions can be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation can also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.


[0161] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.


[0162] In cases where it is desirable to prolong the effect of the nitric oxide mimetic, the absorption of the nitric oxide mimetic from subcutaneous or intramuscular injection can be slowed. This can be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the nitric oxide mimetic then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered formulation is accomplished by dissolving or suspending the nitric oxide mimetic in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations can also be prepared by entrapping the nitric oxide mimetic in liposomes or microemulsions which are compatible with body tissues.


[0163] Formulations for rectal or vaginal administration are preferably suppositories, sponges or rings, which can be prepared by mixing the nitric oxide mimetics with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity thereby releasing the nitric oxide mimetic.


[0164] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the nitric oxide mimetic is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia; humectants such as glycerol; disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents such as paraffin; absorption accelerators such as quaternary ammonium compounds; wetting agents such as cetyl alcohol and glycerol monostearate; absorbents such as kaolin and bentonite clay; and lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.


[0165] Solid compositions of a similar type can also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the nitric oxide mimetic only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.


[0166] Powders and sprays can contain, in addition to the nitric oxide mimetic, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons or alternative non CFC propellants such as DIMEL, also referred to as 1,3,4-A.


[0167] Dosage forms for topical or transdermal administration of nitric oxide mimetics include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The nitric oxide mimetic is admixed under sterile conditions with a pharmaceutically acceptable carrier and any preservatives and/or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention. Transdermal patches have the added advantage of providing controlled delivery of the nitric oxide mimetic to the body. Such dosage forms can be made by dissolving or dispensing a nitric oxide mimetic in the proper medium. Absorption enhancers can also be used to increase the flux of the nitric oxide mimetic across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the nitric oxide mimetic in a polymer matrix or gel.


[0168] The ointments, pastes, creams and gels may contain, in addition to a nitric oxide mimetic, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.


[0169] A preferred mode of delivery is one which provides a reasonably steady-state delivery of the nitric oxide mimetic, so as to maintain steady-state plasma concentrations. Such delivery avoids any substantial initial spike in plasma concentration of the agent, as it would be desirable to avoid plasma concentrations that produce negative side effects. Transdermal patches and pulsed delivery systems are preferred modes of delivery.


[0170] For those formulations containing a nitric oxide mimetic which is commercially available, the low dose formulations for use in the method of the present invention are preferably formulated according to the same methods as the commercially available higher dose formulations, but with lower amounts sufficient to increase, restore or maintain nitric oxide mimetic activity to cells at a level which inhibits or prevents malignant cell phenotypes and/or enhances the efficacy of an antimalignant therapeutic modality. Methods of formulation are within the skill of pharmaceutical formulation chemists and are fully described in such works as Remington's Pharmaceutical Science, 18th Edition, Alfonso R. Gennaro, Ed., Mack Publishing Co., Easton, Pa., USA, 1990.


[0171] The methods and formulations of the present invention are particularly useful in inhibiting metastases and development of resistance of tumor cells to antimalignant therapeutic modalities including, but not limited to chemotherapeutic agents, radiation therapy, immunotherapies, and thermal therapies. Examples of classes of chemotherapeutic agents useful in combination with low dose NO mimetics include, but are not limited to: anti-angiogenic agents including, but not limited to anti-VEGF agents, alkylating agents such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics such as anthracyclines, bleomycins, dauxorubicin, mitomycin, dactinomycin, and plicamycin; endothelin activating agents; enzymes such as L-asparaginase; farnesyl-protein transferase inhibitors; 5α reductase inhibitors; inhibitors of 17β-hydroxy steroid dehydrogenase type 3; hormonal agents such as glucocorticoids, estrogen or antiestrogens, androgens or antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonist; octreotide acetate; microtubule-disruptor agents, such as ecteinascidins and analogs and derivatives thereof; microtubule-stabilizing agents such as taxanes, for example, TAXOL (paclitaxel), TAXOTERE (docetaxel) and thereof analogs, and epothilones or analogs thereof; vinca alkaloids; epipodophyllotoxins; topoisomerase inhibitors; prenyl-protein transferase inhibitors; and other agents such as hydroxyurea, procarbazine, mitotane, hexamethylmelamine, platinum coordination complexes such as cisplatin and carboplatin, biological response modifiers, growth factors, and immune modulators or monoclonal antibodies. Representative examples of chemotherapeutic agents in these classes useful in the present invention include but are not limited to, actinomycin D, aflacon, bleomycin sulfate, buserelin, busulfan, carmustin, chlorambucil, cladribin, cyclophosphamide, cytarabine, dacarbazine, daunorubicin, discodermolides, doxorubicin hydrochloride, estramustine, estramustine phosphate sodium, etoposide, etoposide phosphate, fludarabine, fluorouracil, flutamide, idarubicin, ifosfamide, interferon, interleukins, leuprolide, levamisole, lomustine, mechlorethamine hydrochloride, melphalan, mercaptopurine, methotrexate, mitomycin C, paclitaxel, pentastatin, pteridine, quinocarcins, rituximab, safracins, saframycins, semustine, streptozocin, tamoxifen, teniposide, thioguanine, thiotepa, topotecan, vinblastine, vincristine, vinorelbine tartrate, aldesleukin (Interleukin-2; proleukin), alemtuzumab (Campath; monocloncal antibody against 21-28 kDa cell surface glycoprotein CD52), altretamine (Hexalen; hexamethylmelamine; HMM), amifostine (Ethyol; WR2721), aminoglutethimide (Cytadren; Epithen), amsacrine (m-AMSA;AMSA;Amsidyl), anastrozole (Arimidex), arsenic trioxide (Trisenox), asparaginase (Elspar; L-Asparagionase), bacillus calmette-guerin (BCG; TheraCyst; TICE), bexarotene (Targretin), bicalutamide (Casodex), capacitabine (Xeloda), carmustine (BCNU; Bischloronitrosourea), daunorubicin liposome (DaunoXome), denileukin diftitox (Ontak; DAB389IL-2), dexrazoxane (Zinecard; ICRF-187), docetaxel (Taxotere), doxorubicin (Adria' DOX; Rubex; Adriamycin; hydroxydaunorubicin), doxorubicin liposome (Doxil), epirubicin (Ellence; EPI;), erythropoietin (Procrit; Epogen; EPO), exemestane (Aromastin), filgrastim (Neupogen; G-CSF [granulocyte colony stimulating factor]), floxuridine (FUDR; 5-fluoro-2-deoxyuridine), 5-fluorouracil (5-FU; Efudex), gemcitabine (Gemzar), gemtuzumab ozogamicin (CMA-676; Mylotarg), goserelin (Zoladex), imatinib (ST1570; gleevec; CGP57148B), interferon alpha (Intron A; Roferon), irinotecan (CPT-11; Camptosar), isotretinoin (Accutane; 13-cis-retinoic acid), letrozole (Femara), mechlorethamine (Mustargen), megestrol acetate (Megace), mesna (Mesnex; 2-mercaptoethanesulfonic acid), mitomycin-C (Mutamycin), mitoxantrone (Novantrone; DHAD; DHAQ), nilutamide (Nilandron), oxaliplatin (Eloxatin; DACH-15 platinum), pemetrexed (Alimta; LY231514), pentostatin (dCF; Nipent; 2′-deoxycoformycin), procarbine (Matulane; Natulanar; N-Methylhydrazine), raltitrexed (Tomudex; ZD1694), sargramostim (Leukine; GM-CSF[granulocyte monocyte colony stimulating factor]), streptozocin (Zanosar; Streptozotocin), UFT (Uracil; Tegafur), temozolomide (Temodar), thalidomide (Thalomid), toremifene (Fareston), trastuzumab (Herceptin, Anti-HER-2 antibocy; rhuMAbHER2), tretinoin (Vesanoid; all-trans retinoic acid), trimetrexate (TMTX; Neutrexin; TMQ), vinorelbine (Navelbine), and any analogs or derivatives thereof.


[0172] Animals suffering from cancer can be administered a low dose of a nitric oxide mimetic to inhibit the metastatic potential of the tumor cells as well as to enhance the efficacy of a co-administered antimalignant therapeutic modality targeted at killing the cancer cells. In this embodiment, the nitric oxide mimetic can be administered to animals in combination with other antimalignant therapeutic modalities, following, prior to or during surgical removal or a tumor, and/or following, during, or prior to radiation or thermal therapy. It is believed that this therapy will also enhance the efficacy of anti-VEGF agents targeted at inhibiting angiogenesis of vascular endothelial cells to tumors. In this embodiment, low dose nitric oxide mimetic therapy can be administered to an animal prior to, with, or following administration of an anti-VEGF agent such as anti-VEGF antibody. In this embodiment, it is preferred that the nitric oxide therapy be maintained at least throughout the known active period of the anti-VEGF agent. Low dose nitric oxide mimetic therapy can also be administered as prophylactic therapy to animals at high risk for developing cancer to prevent the development of cells with a malignant cell phenotype. In this embodiment, a low dose of the nitric oxide mimetic may be administered daily to the animal throughout its life. Accordingly, administration of long-term sustained release dosing formulations may be preferred in these animals. In addition, low dose nitric oxide therapy can be administered to animals suspected of, or known to be, exposed to a factor which lowers cellular nitric oxide mimetic activity so as to induce cells with a malignant cell phenotype. Administration of this low dose nitric oxide therapy is expected to inhibit development of a malignant cell phenotype in these animals. In this embodiment, it is preferred that the nitric oxide mimetic therapy be administered for at least as long as the animal is exposed to the factor. For example, both surgery and anesthesia are believed to be factors which lower cellular nitric oxide mimetic activity so as to induce a malignant cell phenotype. Accordingly, prior to or during a surgical procedure and/or administration of an anesthetic agent in an animal, the animal can also be administered a low dose of a nitric oxide mimetic to prevent and inhibit a malignant cell phenotype. In this embodiment, it is preferred that the nitric oxide mimetic be administered for at least the time in which the animal is undergoing the surgical procedure and/or is under the effects of anesthesia. Similarly, an animal subjected to physical trauma, especially a physical trauma associated with blood loss, a decrease in blood volume or hemorrhage can be administered a low dose of a nitric oxide mimetic to prevent and inhibit a malignant cell phenotype. It is believed that co-administration of a low dose of a nitric oxide mimetic can also be used to inhibit or prevent a malignant cell phenotype which may occur upon administration of pharmacological agents which alter the circulation, e.g. antihypertensives. In this embodiment, the nitric oxide mimetic is preferably administered on a daily basis with the other agents or in a long-term sustained release formulation which extends over the period in which the other agent is administered.


[0173] The following nonlimiting examples are provided to further illustrate the present invention.



EXAMPLES


I. Example 1


Materials and Methods

[0174] A. Materials


[0175] Tissue culture medium (RPMI 1640) and fetal bovine serum (FBS) were purchased from Gibco BRL (Grand Island, N.Y.). Hypoxic conditions were generated using airtight chambers from BellCo Biotechnology (Vineland, N.J.). GTN was obtained as a solution (TRIDIL, 5 mg ml−1 or 2.22 M) in ethanol, propylene glycol and water (1:1:1.33) from DuPont Pharmaceuticals (Scarborough, ON). Sodium nitroprusside (SNP) was purchased from Sigma Chemical Co. (St. Louis, Mo). RNA extractions were conducted using a PURESCRIPT RNA isolation kit from Gentra Systems (Minneapolis, Minn.). For the Northern blot analyses, the nylon membranes used for the RNA transfers were purchased from Micron Separations, Inc. (Westboro, Mass.); the uPAR and PAI-1 cDNA probes were cloned in a Bluescript plasmid vector; the [32P]-dCTP and the Reflection NEF film were purchased from Dupont/New England Nuclear (Mississauga, ON); and the oligolabelling kit was obtained from Pharmacia Biotech (Piscataway, N.J.). For the in vitro invasion assays, the serum-free EX-CELL 300 culture medium was purchased from JRH Biosciences (Lenexa, Kans.), the Costar TRANSWELL inserts (6.5 mm diameter polycarbonate, membrane, 8 μm pore) were purchased from Corning Costar (Cambridge, Mass.), and the reconstituted basement membrane (MATRIGEL) was bought from Collaborative Biomedical Products (Bedford, Mass.). The plasminogen activator inhibitor-1 (PAI-1) enzyme-linked immunosorbent assay (ELISA) kit was obtained from American Diagnostica (Greenwich, Conn.). For the Western blot analysis of uPAR, the resolved proteins were transferred to lmmobilon-P membranes from Millipore (Bedford, Mass.), anti-uPAR antibody (monoclonal antibody [MoAb] 3937) was purchased from American Diagnostica (Greenwich, Conn.), the blotting grade affinity purified goat anti-mouse IgG (H+L) horseradish peroxidase conjugate was obtained from BIO-RAD (Hercule Calif.), and the antigen was detected by enhanced chemiluminescence (ECL) using reagents from Amersham Canada (Mississauga ON). For the zymographic analyses, the gelatin was purchased from BDH (Toronto, ON), the casein was bought from Sigma Chemical Co. (St. Louis, Mo.) and the plasminogen was from American Diagnostica (Greenwich, Conn.).


[0176] B. Cells


[0177] The HTR-8/SVneo invasive trophoblast cell line and the MDA-MB-231 metastatic breast carcinoma cell line were used in these experiments. Both the HTR-8/SVneo and the MDA-MB-231 cells were cultured in RPMI-1640 medium supplemented with 5% FBS.


[0178] The HTR-8/SVneo cell line was obtained from explant cultures of human first trimester placenta and immortalized by transfection with a cDNA construct encoding the SVneo large T antigen. These cells have been previously characterized and have been maintained in culture for over 130 passages in RPMI 1640 medium supplemented with 5% FBS. They exhibit a high proliferation index and share various phenotypic similarities with the non-transfected parent HTR-8 cells such as in vitro invasive ability and lack of tumorigenicity in nude mice.


[0179] The MDA-MB-231 cell line was initially isolated in 1973 from the pleural effusion of a 51-year-old breast cancer patient (Callieau et al. J. Nat. Cancer Inst. 1974 53:661-674).


[0180] C. Hypoxic Cell Culture Conditions


[0181] Cells were placed in an airtight chamber and were flushed with a gas mixture containing 5% CO2:95% N2 until the oxygen concentration was 0%, as read by a Miniox 1 Oxygen Analyzer (Catalyst Research Corp., Owing Mills, Md.). The cells were then incubated at 37° C. Within the first 2 hours of incubation, the oxygen level in the chambers had equilibrated to approximately 1%, and remained at this level for the remainder of the incubation period.


[0182] Alternatively, cells were placed in a chamber in which atmospheric O2 levels were maintained by a PRO-OX O2 regulator (Biospherix, Redfield, N.Y.).


[0183] D. Treatment of Cells with Glyceryl Trinitrate (GTN) and Sodium Nitroprusside (SNP)


[0184] In these experiments, the cells were treated with varying concentrations of GTN and SNP. The stock solution of GTN was first diluted in phosphate-buffered saline (PBS) to a concentration of 10−4 M. Following filtration, the GTN solution was diluted in the culture medium to concentrations ranging from 10−4 M to 10−11 M. The SNP (originally in crystal form) was dissolved in distilled water and diluted to a concentration of 10−5 M. Following filtration, the SNP was diluted in the culture medium to concentrations ranging from 10−6 M, to 10−12 M.


[0185] E. Northern Blot Analyses


[0186] Cells were cultured with varying concentrations of GTN and SNP under hypoxic (1% O2) or control (20% O2) conditions at 37° C. In another set of experiments MDA-MB-231 cells were incubated in the presence or absence of LMMA (0.5 mM) for 24 hours under various levels of oxygen (1%, 5% or 20% O2) at 37° C. Following incubation, the total cellular RNA from the cells was isolated using the Gentra PURESCRIPT RNA Isolation Kit. The isolated RNA was subsequently separated by electrophoresis, and transferred to charged nylon membranes. The membranes were prehybridized at 42° C. for approximately 2 hours in a solution containing 50% formamide, 5×Denhardt's solution, 0.5% sodium dodecyl sulfate (SDS), 6×SSC (1×SSC=0.15 M NaCl, 15 mM sodium citrate, pH 7.0) and 100 μg/mL denatured salmon sperm DNA. They were then hybridized with a [32P]-dCTP-labeled cDNA probe (uPAR or PAI-1) for approximately 24 hours at 42° C. in a solution composed of 6×SSC, 0.5% SDS, 100 μg/mL denatured salmon sperm DNA and 50% formamide, and containing a cDNA probe (uPAR or PAI-1) which was labeled with [32P]-dCTP using a Pharmacia Oligolabelling kit. Following serial washes, the membrane was used to expose Dupont Reflection NEF film. After 1-4 days, the film was developed and analyzed. To correct for differences in the amount of RNA loaded in each sample, 28S rRNA was used.


[0187] F. Western Blot Analysis of uPAR Protein Levels


[0188] To examine the level of uPAR protein, the cells were first cultured with 20% O2 or 1% O2 in the presence of varying concentrations of SNP or GTN. Following incubations, the cells were lysed using a buffer containing 40 mM HEPES pH 7.2, 100 mM NaCl, 20% glycerol, 0.1 mM EDTA pH 8.0, 0.2% Triton X-100, 1 mM DTT, and 2 mM PMSF. The lysates were then subjected to homogenization, DNA shearing (10 times with a 25⅝-gauge needle), boiling (5 minutes) and centrifugation (15 minutes, 14000 g). The supernatant was collected and stored at −80° C. until use. The samples were subjected to SDS-polyacrylamide gel electrophoresis (PAGE) and the resolved proteins were transferred to an lmmobilon-P membrane using a wet transfer apparatus (Bio-Rad Laboratories, Mississauga, ON). The membranes were blocked overnight at 4° C. in a solution containing 1% PBS, and 0.01% Tween20 (PBS-T) as well as 5% casein. The blots were subsequently incubated for 1.5 hours with the monoclonal anti-uPAR antibody [MoAb 3937], followed by six 5 minute washes with PBS-T. The membranes were then incubated with a horseradish peroxidase labeled goat anti-mouse IgG secondary antibody for 1.5 hours. Following six additional 5 minute washes with PBS-T, the antigen was detected by enhanced chemiluminescence and the blots were exposed onto Dupont Reflection NEF film.


[0189] G. Measurement of Metalloproteinase and Plasminogen Activator Activity by Zymography


[0190] To measure the levels of metalloproteinase and plasminogen activator activity, the cells were incubated under hypoxic or control conditions in the presence of varying concentrations of SNP or GTN. The cells were cultured using a serum-free medium (EX-CELL 300). Following incubation, the medium was extracted and centrifuged at 5,000 RPM for 5 minutes. The supernatant was subsequently collected and stored at −80° C. until further use. SDS-polyacrylamide gels were prepared in accordance with well-known procedures. For the analysis of metalloproteinase secretion, the gel contained 0.1% w/v gelatin, and for the analysis of plasminogen activator secretion, the gel was supplemented with 0.1% w/v casein as well as plasminogen (50 μg/mL). The serum-free conditioned medium was then combined with a nonreducing sample loading buffer (0.5 M Tris, 10% SDS, 1% Bromophenol Blue in 2 mL glycerol) in a ratio of 4:1 and was not boiled. Following electrophoresis the gels underwent two 15 minute washes with 2.5%. Triton X-100. This step removed the SDS. After the washes with H2O, the gels were incubated overnight at 37° C. in a solution containing Tris-HCl (pH 7.0) and CaCl2 (5 mM). The gels were stained with 0.4% Coomassie Brilliant Blue R-250 in 40% methanol/10% glacial acetic acid/50% distilled water for approximately 1 hour, and then destained for about 2 hours in 30% methanol/10% glacial acetic acid/60% distilled water. Molecular weight standards showed as dark bands against the lighter blue background and colorless zones appeared where lysis occurred. In the gelatin-containing gels, these areas corresponded to metalloproteinase (gelatinase) activity and in the casein gels, these bands corresponded to plasminogen activator activity. The gels were preserved using a preserving solution (10% glacial acetic acid/10% glycerol/80% distilled water) for 1 hour and were dried on cellophane for 1 hour at 60° C.


[0191] H. In Vitro Invasion Assays


[0192] Matrigel invasion chambers (modified Boyden chambers) were used to assess the invasive ability of the cells under hypoxic and standard conditions in the presence or absence of various concentrations of GTN or SNP. The chambers consist of cell culture inserts, 6.5 mm in diameter and with a 8 μm pore size membrane. Each membrane was coated with 100 μL of a 1 mg/mL solution of Matrigel diluted in cold serum-free culture medium (EX-CELL 300), and allowed to dry in a laminar flow cabinet for approximately 12 hours. The Matrigel was then rehydrated by incubating it with 100 μL of serum-free medium for approximately 1 hour. After rehydration, cell suspensions containing 5.0×104 or 1.0×105 cells in 100 μL of medium, containing both serum and the nitric oxide treatments, were added to each well. Culture medium containing serum and the respective nitric oxide treatment were then added to each insert. The cells were incubated for 24 hours under either hypoxic (1% O2) or control (20% O2) conditions. Following incubation, the non-invading cells were removed from the upper surface of the membrane by wiping with a cotton swab. The cells on the lower surface of the membrane were fixed for 10 minutes with Carnoy's fixative (25% acetic acid, 75% methanol), and then stained for approximately 3 hours with a 1% toluidine blue, 1% sodium borate solution. Following a rinse in phosphate-buffered saline (PBS), the membrane was removed from the insert housing with a small scalpel blade, mounted onto a microscope slide and coverslipped. Invading cells were then viewed under the microscope at 40×magnification and counted. The invasion index for each treatment was calculated by dividing the number of invading cells by the number of cells which invaded under standard conditions. This value was then multiplied by 100 to obtain a percentage. The standard was given a value of 100% and the treatment values were converted to a percentage of the standard. The results were tested for statistical significance using either the Tukey test for pair-wise multiple comparison procedures or the Student-Newman-Keuls method for pair-wise multiple comparison procedures. See FIG. 1.


[0193] I. In Vivo Metastasis Model


[0194] C57BL6 mice were injected i.v. (tail vein) with abolus of5×104-105 B16F10 metastatic melanoma cells. Immediately after the tail vein injection, mice were randomly divided into groups of 15 and mice in each group were placed in plexiglass chambers (approximately 3 L) which were continuously flushed with gas mixtures of 20% O2:balance N2 and 10% O2:balance N2, respectively. Gas flows were adjusted to a level which did not allow CO2 build-up within the chambers. After a 24 hour exposure to an atmosphere of either 20% O2 or 10% O2, mice were removed from the chambers and placed in regular cages kept at room air. Thirteen days later, mice were sacrificed by cervical dislocation, and lungs were removed and fixed in Bouin's fixative (Sigma). Metastatic nodules (many of which appeared black due to the presence of melanin) on the surface of the lungs were counted visually under a dissecting microscope. Data were expressed as the number of lung nodules per 104 cells injected and were analyzed using statistical tests for non-parametric values.


[0195] In a second set of experiments, the same protocol was followed except that the B16F10 mouse melanoma cells were incubated for 12 hours in 1% or 20% oxygen in the presence or absence of 2×10−11 M GTN. Cells were then removed from plates with trypsin and 5×104 cells were injected i.v. (tail vein) into C57BL6 female mice. See FIG. 2. Some of the cells treated in vitro were plated onto tissue culture dishes to determine colony-forming ability using the protocol described below in the Section I.J.


[0196] J. Colony Formation Assay for Doxorubicin Resistance


[0197] The resistance of MDA-MB-231 breast cancer cells to doxorubicin was determined following culture in 20% or 1% oxygen by counting the number of colonies formed. MDA-MB-231 cells were incubated in 1% O2 or 20% O2 for 24 hours. Following incubation, the cells were exposed to diluent (control), 25 μM doxorubicin, 25 μM doxorubicin plus 10−6 M GTN or 25 μM doxorubicin plus 10−10 M GTN for 1 hour. The cells were washed and then plated onto 35 mm plates at different dilutions. The cells were incubated for an additional 1-2 weeks in order to allow cell colonies to grow. At the end of the incubation period, the cells were fixed with Carnoy's fixative, stained with Crystal violet, rinsed and allowed to air dry. Colonies were counted visually. The surviving cells under each condition was determined by counting the number of colonies and was expressed as a fraction of the number of colonies that survived without doxorubicin exposure.



II. Example 2


The Activation of sGC and PKG is Linked to the Regulation of Invasive and Metastatic Phenotypes

[0198] A. Materials


[0199] Glyceryl Trinitrate (GTN) at a concentration of 2.22 M (Sabex, Boucherville, QC) and Diethylenetriamine/nitric oxide adduct (DETA-NONOate) (SIGMA Chemical Co., St. Louis, Mo.) were used as NO-mimetics. Desferoxamine mesylate (DFO) (SIGMA Chemical Co., St. Louis, Mo.) was used as an iron chelator. 1H-[1,2,4]Oxadiazolo-[4,3-a]Quinoxalin-1-One (ODQ) (SIGMA Chemical Co., St. Louis, Mo.), was used to selectively inhibit soluble guanylyl cyclase. Various concentrations 8-bromoguanosine 3′:5′-cyclic monophosphate (8-Br-cGMP) (SIGMA Chemical Co., St. Louis, Mo.) were used to activate cGMP-dependent protein kinase (PKG), and KT5823 (Calbiochem, La Jolla, Calif.) was used to selectively inhibit PKG.


[0200] B. Cells


[0201] The HTR-8/SVneo line was established from explant cultures of human first trimester placenta and was immortalized by transfection with the large T antigen of simian virus 40. These cells exhibit similar in vitro invasive abilities to their parent HTR-8 cells, and are not tumourigenic in nude mice. They have been maintained in culture for over 130 passages, and require at least 5% serum in order to sustain growth in vitro (see, Graham, Hawley, et al. 1993 ID: 1080).


[0202] The MDA-MB-231 is a metastatic breast cancer cell line that was initially isolated in 1973 from a 51-year-old patient {Cailleau, Young, et al. 1974 ID: 982}.


[0203] A metastatic murine melanoma (B16F10) (a kind gift of Dr. Ann Chambers, London Regional Cancer Centre, London, ON) was utilized in the experimental metastasis experiments.


[0204] All of the cell cultures were maintained in monolayer culture in RPMI 1640 medium (Gibco BRL, Grand Island, N.Y.) supplemented with 5% fetal bovine serum (FBS; Gibco BRL) in a standard Sanyo CO2 incubator (5% CO2 in air, 37° C.; Esbe Scientific, Markham, ON).


[0205] C. Culture Conditions


[0206] In order to establish hypoxic culture conditions, cells were placed in airtight chambers (BellCo Biotechnology, Vineland, N.J.) which were flushed with a gas mixture of 5% CO2/95% N2 until the desired oxygen concentration was reached. Oxygen levels were maintained through the use of a Pro-Ox Model 110 oxygen regulator (Reming Biospherix, Redfield, N.Y.).


[0207] D. Northern Blot Analysis


[0208] Following incubation, the total cellular RNA was isolated using a GENTRA PURESCRIPT® RNA Isolation Kit (Gentra Systems, Minneapolis, Minn.). The isolated RNA was subsequently separated by electrophoresis, transferred to a charged nylon membrane (Micron Separations Inc., Westboro, Mass.) and fixed with ultraviolet radiation using a UV Crosslinker (NAME). The membranes were prehybridized at 68° C. for approximately 1 hour in a prewarmed ultrasensitive hybridization buffer, ULTRAhyb™ (Ambion, Austin, Tex.). They were then hybridized overnight at 68° C. with a DIG-labeled uPAR probe. In order to generate the probe, a uPAR cDNA cloned in a Bluescript plasmid vector was linearized. The linear cDNA subsequently underwent in vitro transcription with a DIG-labelled RNA labeling mix and T3 RNA polymerase (Roche Molecular Biochemicals, Indianapolis, Ind.). Following hybridization, the membranes were washed twice in 2×SSC, 0.1% SDS (5 minutes at 68° C.) and then twice in 0.1×SSC, 0.1% SDS (15 minutes at 68° C.). Bands were detected using a DIG luminescent detection kit (Roche Molecular Biochemicals, Indianapolis, Ind.) and the membrane was exposed to Dupont Reflection NEF film (Dupont/New England Nuclear, Mississauga, ON).


[0209] E. Western Blot Analysis


[0210] Following incubation, the cells were lysed using a buffer containing 40 mM HEPES pH 7.2, 100 mM NaCl, 20% glycerol, 0.1 mM EDTA pH 8.0, 0.2% Triton X-100, 1 mM dithiothreitol (DTT), and 2 mM phenylmethyl sulfonyl fluoride (PMSF). The lysates were then subjected to homogenization, DNA shearing (10 times with a 25⅝-gauge needle), boiling (5 minutes) and centrifugation (15 minutes at 14,000×g). The supernatant was collected and stored at −80° C. until use. The samples were subjected to SDS-polyacrylamide gel electrophoresis (PAGE) and the resolved proteins were transferred to an lmmobilon-P membrane (Millipore Corp., Bedford, Mass.) using a wet transfer apparatus (BioRad Laboratories, Mississauga, ON). The membranes were blocked overnight at 4° C. in a solution containing 1% PBS, and 0.01% Tween 20 (PBS-T) as well as 5% dry milk powder. The blots were subsequently incubated for 1.5 hours with the monoclonal anti-uPAR (MoAb 3937; American Diagnostica Inc., Greenwich, Conn.), or a polyclonal soluble guanylate cyclase (sGC) antiserum (Cayman Chemical, Ann Arbor, Mich.) followed by six 5-minute washes with PBS-T. Subsequently, the uPAR membranes were incubated for 1 hour with a horseradish peroxidase labelled goat anti-mouse IgG secondary antibody (Bio-Rad, Hercules, Calif.) and the sGC membranes were incubated for 1 hour with a goat anti-rabbit IgG horseradish peroxidase conjugate (VECTOR Laboratories Inc., Burlingame, Calif.). Following six additional 5-minute washes with PBS-T, the antigen was detected by enhanced chemiluminescence (Amersham Canada Inc., Mississauga, ON) and the blots were exposed onto Dupont Reflection NEF film.


[0211] F. In Vitro Invasion Assays


[0212] Matrigel invasion assays were used to assess the effect of 8-Br-cGMP on the ability of cells to penetrate through an extracellular matrix. The chambers consisted of 6.5-mm inserts with 8-μm pore membranes (Corning Costar Corp., Carmbridge, Mass.). Each membrane was coated with 100 μL of a 2.6 mg/mL solution of Matrigel (Collaborative Biochemical Products, Bedford, Mass.) diluted in cold serum-free medium (EX-CELL 300; JRH Biosciences, Lenexa, Kans.), and allowed to dry in a laminar flow cabinet for approximately 12 hours. The Matrigel was then rehydrated with 100 μL of serum-free medium for approximately 1 hour. After rehydration, cell suspensions containing 5.0×104 cells in 100 μL of RPMI, as well as serum (5% FBS) and 8-Br-cGMP, were added to each well. The cells were incubated for 24 hours in either 1% or 20% oxygen. Following incubation, the non-invading cells were removed from the upper surface of the membrane by wiping with cotton tipped swabs. The cells on the lower surface of the membrane were fixed for 2 minutes with ice-cold methanol, and stained overnight with a 1% toluidine blue, 1% sodium borate solution. Following a rinse in phosphate-buffered saline (PBS), the membrane was removed from the insert with a small scalpel blade, mounted on a microscope slide and cover-slipped. Invading cells were then viewed under the microscope at 40×magnification and counted. The invasion index for each treatment was calculated by dividing the number of invading cells by the number of cells that invaded under standard (20% oxygen) conditions. Invasion indices were expressed as percentages of control (20% oxygen) values. The results were tested for statistical significance using analysis of variance followed by either the Tukey or the Student-Newman-Keels tests for pair wise multiple comparisons.


[0213] G. In Vivo Metastasis Experiment


[0214] A murine metastasis model was used to assess the effect of 8-Br-cGMP on the metastatic potential of B16-F10 melanoma cells. The cells were incubated for 12 hrs in 1% or 20% oxygen and in the presence or absence of 8-Br-cGMP (1 μM). Following harvesting from culture plates, 5×104 cells were injected intravenously (tail vein injections) into 5-7 week old C57B1/6 female mice. Cells were also plated in tissue culture plates to determine the effect of culture conditions on in vitro colony forming ability. Fourteen days after intravenous inoculation, mice were sacrificed and their lungs were removed and fixed in Bouin's fixative. Both melanotic and amelanotic metastatic colonies were counted under a dissecting microscope. Data were expressed as the number of lung nodules per 5×104 viable cells injected (as determined by in vitro colony-forming ability) and were analyzed using statistical tests for non-parametric values.


[0215] H. Measurement of Guanylate Cyclase Levels and Activity


[0216] The effects of hypoxia and NO-mimetic treatment on sGC levels were determined using Western Blot analysis. To determine sGC activity, cGMP levels were measured. Briefly, cells were extracted over ice in 1 mL of 6% trichloroacetic acid (BDH Laboratory Supplies, Poole, England). The homogenate was then centrifuged at 13,000 g for 10 min. The supernatant fraction was removed and extracted five times with 2 mL of water-saturated diethyl ether (BDH). The cGMP contained in this fraction was subsequently acetylated and quantitated using an enzyme-linked immuno-sorbent assay (ELISA) kit (STI—Signal Transduction Products, San Clemente, Calif).


[0217] Results


[0218] I. The Role of Soluble Guanylate Cyclase in the NO Mimetic Prevention of Hypoxia Induced uPAR Upregulation


[0219] To determine whether a heme protein, such as sGC played a role in the ability of NO mimetics to inhibit the hypoxic upregulation of uPAR, the iron chelator Desferroxamine (DFO) (100 μM ) was used. It was found that the upregulation of uPAR that occurred in response to heme disruption with DFO could not be inhibited by GTN (10−10 M, 10−6 M). These results suggest that the NO-mimetic effect is mediated via a heme moiety.


[0220] Western blot analysis was used to determine the effects of various oxygen concentrations as well as NOS blockade on sGC protein levels. It was found that the total sGC levels remained unchanged, regardless of the treatment employed. Conversely, using an ELISA for cGMP it was found that sGC activity is decreased after only 2 hours of hypoxia (0.5% O2). Further, this reduction was significantly prevented by the addition of GTN (10−10 M, 10 −6 M).


[0221] The selective sGC inhibitor ODQ (0.5 μM) was used to determine the role of sGC in the NO mimetic prevention of uPAR upregulation. It was found that the ability of GTN (10−6 M) to abrogate the hypoxic induction of uPAR was substantially prevented by ODQ at both the protein and mRNA levels.


[0222] J. The Role of PKG in the NO Mimetic Prevention of Hypoxia Induced uPAR Upregulation


[0223] To determine the role of PKG in the in the oxygen mediated control of uPAR, the PKG activator 8-Br-cGMP (10−3-10−7 M) was added to cells cultured in 20 and 0.5% O2 or in the presence of DFO (100 μM). In accordance to previous studies, uPAR expression was markedly increased under hypoxic conditions and in the presence of DFO at both the protein and mRNA levels. Interestingly, this induction was significantly abrogated by the addition of 8-Br-cGMP. To assure that the 8-Br-cGMP was activating PKG, a PKG inhibitor (KT5823, 10 μM) was added to the medium. It was found that KT5823 could induce the expression of uPAR, even in the presence of oxygen and that it significantly blunted the effects of 8-Br-cGMP.


[0224] K. The Effects of 8-Br-cGMP on Hypoxia-Induced Invasion and Metastasis


[0225] As a functional correlate of uPAR expression, an in vitro invasion assay was conducted. It was found that 8-Br-cGMP could significantly inhibit hypoxia-associated invasion.


[0226] To extend these findings, an in vivo metastasis assay was conducted. In accordance to previous studies, culture of the B16F10 cells in hypoxic conditions (1% O2) resulted in a marked increase in metastatic lung nodules. This induction was completely abrogated by the addition of 8-Br-cGMP (1 μM). Using a colony formation assay it was found that the 8-Br-cGMP treatment did not affect the viability of the cells.


[0227] L. The Effects of NO Mimetics on the Hypoxic Upregulation of In Vitro Invasion and Expression of uPAR


[0228] To determine the effect of NO mimetics on hypoxia during in vitro invasiveness, an in vitro invasion assay was conducted. While hypoxia increased the in vitro invasiveness of MDA-MB-231 breast carcinoma cells by more than 5-fold (FIG. 5), it was found that low concentrations of two different NO-mimetic drugs, glyceryl trinitrate (GTN; 1 pM and 0.1 μM; P<0.002 and P<0.001 respectively; one-way ANOVA followed by Fisher's test) and sodium nitroprusside (SNP; 10 pM; P<0.001), administered at the beginning of the 24-hour invasion assay.


[0229] MDA-MB-231 cells were cultured in 0.5% oxygen for 24 hours to determine the effect on uPAR protein levels. It was found that exposure to 0.5% oxygen resulted in up to 3.5-fold increases (P<0.0001) in uPAR protein levels (FIGS. 6A and 9B) and up to 5-fold increases (P<0.0002) in uPAR mRNA levels (FIGS. 6C, 8A, and 9A). Administration of single doses of GTN (1 pM, 1 nM, and 0.1 μM) to cells incubated for 24 hours in 0.5% oxygen was sufficient to prevent the upregulation of uPAR protein expression (FIG. 6A; P<0.004, <0.005 and <0.001 for each concentration of GTN, respectively). Similarly, the hypoxia-mediated increase in uPAR mRNA levels was also inhibited when a low concentration of GTN (a pM) was used (FIG. 6C; P<0.001).


[0230] M. The Effects of NO Synthase Inhibition on uPAR Protein and mRNA Levels


[0231] To assess whether endogenous NO inhibits uPAR expression, NO synthesis in MDA-MB-231 cells was blocked by incubation with the NOS inhibitor N-monomethyl-L-arginine (L-NMMA, 0.5 mM). In a manner characteristic of cells exposed to hypoxia, a 24-h incubation with a single dose of L-NMMA resulted in an overall 50% increase in the levels of uPAR protein (FIG. 6B; P<0.004) and a 2.8-fold increase in uPAR mRNA levels in MDA-MB-231 cells even when cultured in 20% O2 (FIG. 6D; P<0.04). In contrast, compared with uPAR expression in cells incubated in 20% O2 alone, no significant increase in uPAR protein (P=0.89) or transcript (P=0.35) levels was observed in cells incubated with a combination of L-NMMA (0.5 mM) and GTN (0.1 nM) in 20% O2 (FIGS. 6B and 6D).


[0232] N. The Effects of Hypoxia on sGC Signaling


[0233] To determine the effect of hypoxia on cGMP levels, MDA-MB-231 breast carcinoma cells were incubated in 0.5% O2 for 6 hours in the presence of IBMX (500 μM). The results from an ELISA for cGMP revealed that, compared with cells incubated in 20% O2, the MDA-MB-231 cells incubated in 0.5% O2 exhibited a 50% reduction in accumulated cGMP levels (P<0.002; FIG. 7A). This effect of hypoxia on cGMP levels was prevented by co-incubation with the NO mimetic GTN (1 μM).


[0234] Western Blot analysis was conducted to determine the effects of hypoxia on sGC protein levels. Soluble GC is a heterodimeric protein consisting of an α1 and a β1 subunit. Culture in hypoxia for 24 hours resulted in a 44% decrease in the levels of the β1 subunit (P<0.0001). In contrast, culture in hypoxia resulted in a 2.3-fold increase (P<0.02) in the levels of the a1 subunit (FIG. 7B). Although the ratio of the α1 and the β1 subunits was altered during hypoxia, the total amount of sGC was not significantly changed.


[0235] O. The Role of sGC in the NO-Mediated Inhibition of uPAR Expression


[0236] To determine the effect of sGC blocker on the levels of uPAR mRNA, MDA-MB-231 cells were incubated for 24 hours with the selective sGC blocker (ODQ, 0.5 μM). Compared with incubation of cells under control conditions (20% O2 alone), the treatment described above resulted in a 2.7-fold increase (p<0.05) in the levels of uPAR mRNA (FIG. 8A). While the presence of GTN (1 μM) prevented the hypoxic upregulation of uPAR mRNA expression, GTN was unable to block the effect of hypoxia when ODQ was also present in the medium (FIG. 8A).


[0237] Soluble GC is a heme-containing enzyme that requires ferrous iron for its biosynthesis and activity. Therefore, to further assess the participation of this enzyme in the regulation of uPAR expression by the NO signaling pathway, MDA-MB-231 cells were cultured in the presence of DFO, which resulted in a 4-fold increase (P<0.007) in the levels of uPAR mRNA (FIG. 8C). In contrast to hypoxia, the effect of DFO on uPAR mRNA expression was not prevented by 1 μM of GTN.


[0238] Furthermore, the upregulation of uPAR mRNA and protein expression by hypoxia was significantly reduced (P<0.0001) in a dose-dependent manner (up to 100%) by the presence of 8-Br-cGMP (0.1 μM-10 μM; 24 hours), a non-hydrolysable analogue of cGMP (FIGS. 9A and 9B). Interestingly, the presence of 8-Br-cGMP (1 mM) also resulted in the complete inhibition (P<0.01) of the DFO-induced upregulation of uPAR mRNA expression (FIG. 9C). These results indicate that a major component of the hypoxia- and DFO-mediated stimulation of uPAR expression is the inhibition of sGC.


[0239] P. Role of PKG in the NO Mediated Inhibition of uPAR Expression


[0240] To further elucidate the role of NO signaling in the regulation of uPAR expression, MDA-MB-231 cells were incubated for 6 hours with the PKG inhibitor KT5823 (10 μM) Nortbern and Western blot analyses revealed that selective inhibition of PKG, even in 20% O2, resulted in a 1.8-fold (P<0.05) increase in uPAR protein and mRNA expression (FIGS. 10A and 10B). These results demonstrate that PKG activation by cGMP is necessary for the inhibition of uPAR expression by NO.


[0241] Q. Effect of 8-Br-cGMP on Hypoxia-Induced Invasion


[0242] To determine the effect of 8-Br-cGMP on hypoxia-induced invasion, an in vitro invasion assay was conducted. Results from the in vitro invasion assay using matrigel as a substrate for invasion revealed that, compared to the invasiveness of cells incubated in 20% O2, hypoxia stimulated the invasiveness of MDA-MB-213 cells by 3.9 fold (P<0.0001; FIG. 11). This effect of hypoxia on invasiveness was completely inhibited by the presence of various concentrations of 9-Br-cGMP (0.1 μM-1 mM; FIG. 11).



III. Example 3


The Role of Nitric Oxide in Hypoxia-Induced Chemoresistance in Prostate Cancer Cells

[0243] Low tumor oxygenation (hypoxia) correlates with increased metastasis and resistance to radiotherapy and chemotherapy. Hypoxia has been shown to be associated with human prostate cancer, and prostate cancers are often highly resistant to chemotherapy. To test whether hypoxia is a major contributor to reduced chemosensitivity in prostate cancer cells and that hypoxia-induced chemoresistance in prostate cancer cells can be inhibited by low concentrations of nitric oxide (NO) mimetic agents, human PC-3 and mouse TRAMP-C2 prostatic adenocarcinoma cells were incubated in 20% O2 or 0.5% O2 for 12 hours in the presence or absence of glyceryl trinitrate (GTN, 0.1 nM). This was followed by a one-hour incubation with doxorubicin and survival was assessed by colony formation assays. Western blot analysis was used to measure NO synthase (NOS) protein.levels in cells cultured in 20% and 0.5% O2. Pre-incubation of the two cell lines under hypoxia resulted in increased survival (up to 8-fold for the PC-3 cells and up to 12-fold for the TRAMP-C2 cells; P<0.0001) following exposure to doxorubicin. Co-incubation of the PC-3 and TRAMP-C2 cells with 1 nM of GTN resulted in complete inhibition of the hypoxia-induced resistance to doxorubicin (P<0.0001). Both cell lines expressed all three NOS isoforms (NOS I, NOS II and NOS III), but only the expression of NOS III was significantly increased (P<0.05) in PC-3 cells incubated in 0.5% versus 20% O2. These findings indicate that NO plays an important role in the regulation of chemosensitivity in prostate cancer cells. Furthermore, the results indicate that administration of GTN provides a means of chemosensitizing prostatic carcinomas (FIGS. 12 and 13).



IV. Example 4


Multicellular Resistance to Doxorubicin is Attenuated by Nitric Oxide Mimetics

[0244] Previous studies revealed that hypoxia-induced drug resistance in tumor cells cultured in monolayer results from impaired nitric oxide (NO) production. It is also well established that, compared with culture in monolayer, tumor cells cultured as multicellular aggregates (spheroids) exhibit significantly higher levels of resistance to chemotherapeutic agents. The present study demonstrated that chemoresistance associated with spheroid culture is due to decreased NO signalling. Using human MDA-MB-231 breast carcinoma cells in clonogenic assays, survival was determined following doxorubicin exposure and NO mimetic administration to both monolayer and spheroid cultures. Compared with incubation in 20% O2, monolayer cells incubated in 1% O2 exhibited a three-fold increase in resistance to doxorubicin. Administration of glyceryl trinitrate (GTN), at 1 μM, resulted in a 50% attenuation of resistance. Incubation with 200 μM doxorubicin resulted in a 90% decrease in survival of tumor cells cultured as spheroids. Diethylenetriamine/nitric oxide adduct (DETA) at 0.1 mM and 2 μM, and GTN at 0.1 μnM and 1 μM, did not affect the survival of control (doxorubicin-untreated) cells. However, these NO mimetics effectively attenuated the resistance to 200 μM doxorubicin associated with 3D culture by 90% and 30% respectively. These results demonstrate that treatment with NO mimetics can be used as a novel approach to increase the chemosensitivity of solid tumors (FIGS. 14 and 15).



V. Example 5


The Role of cAMP Signaling in the Regulation of Breast Carcinoma Invasiveness

[0245] This example demonstrates that the hypoxia-induced increases in uPAR expression and invasion are mediated via a reduction in cGMP-dependent signaling. The cGMP and cAMP systems are intimately connected and cGMP levels have been shown to modulate cAMP levels. Given the importance of cGMP signaling in the regulation of invasiveness and its ability to modulate cAMP signaling, this study demonstrates the role of cAMP and its effector, cAMP-dependent protein kinase (PKA), in the regulation of carcinoma cell invasiveness.


[0246] Human breast carcinoma (MDA-MB-231) cells were cultured for 24 hours in 20% or 0.5% O2 in the presence or absence of the adenylyl cyclase activator forskolin (10 μM) and/or the PKA inhibitor H-89 (1 μM). Northern and Western blot analyses showed that forskolin inhibited uPAR mRNA and protein expression at both 20% and 0.5% O2. This effect of forskolin on uPAR expression was prevented by H-89, demonstrating that the effects of forskolin are mediated by the cAMP-dependent activation of PKA. Inhibition of PKA with H-89 also resulted in an up-regulation of uPAR expression that was independent of oxygenation. Using an in vitro invasion assay through reconstituted extracellular matrix (Matrigel®), forskolin inhibited cellular invasion at both 20% and 0.5% O2. Inhibition of PKA activity with H-89 prevented this effect of forskolin and independently enhanced invasiveness. These findings demonstrate that cAMP-dependent signaling plays an important role in the regulation of carcinoma cell invasiveness, independently of oxygenation status. Furthermore, the cAMP pathway provides a novel pharmacological target for the inhibition of tumor cell invasion and metastasis.



VI. Example 6


Low Dose Nitroglycerin Treatment Decreases PSA Velocity

[0247] Methods:


[0248] Patients with a previous history of prostatectomy that have experienced biochemical failure as per the ASTRO criteria (three successive rises in PSA), PSA<10 (units), stable with sub-clinical disease were included in the study after signing an informed consent. All patients were placed on treatment with continuous low dose transdermal nitroglycerin (˜0.03 mg/hr, 24 hours a day). Subjects returned to the study centre for monthly PSA measurements. Subjects were withdrawn from the study if there was evidence of disease progression or other intervening medical events.


[0249] Results: In general patients having 5 or more months of therapy have decreased mean PSA velocity from 14%/month to 5.4%/month, 4 of 9 subjects have completed 6 months of therapy. 1 subject (with negative margins) has experienced a decrease in PSA, the remaining 3 subjects (with positive margins) have experienced a decrease in PSA velocity, 3 subjects were withdrawn due to: initiation of cardiac medication, renal failure and MRI positive for spinal canal metastasis identified just after enrollment. No patients experienced drug related adverse events.


[0250] Conclusions: The current ongoing pilot open-label study indicates that the use of low dose nitric oxide donor treatment is safe and well tolerated in subjects that have experienced biochemical failure after radial prostatectomy. A benign treatment that provides a decrease in PSA velocity during the early phase of biochemical failure can provide a valuable adjunctive clinical tool in the management of prostate cancer. Based on the current data, the concept of low dose nitric oxide treatment of rising PSA associated with biochemical failure should be assessed in a randomized, placebo controlled clinical study.



VII. Example 7


Preventing the Progression of Cervical Intralesional Neoplasia (CIN) to Cervical Cancer

[0251] Cervial cancer is most common in developing countries. The cervix is the lower third of the uterus. Cancer of the cervix may originate on the vaginal surface or in the canal. Each year, an estimated 500,000 new cases are diagnosed worldwide with some 250,000 of those women destined to die of their disease. Ample evidence exists to show that both incidence and mortality can be reduced by the use of cervical screening program (Cain J M, et al, Science, 288:1753-1754, 2000). Currently, human papillomavirus (HPV), especially HPVs 16 & 18, has been implicated as the major causal agent in this disease. Symptoms of cervical cancer include vaginal bleeding, post-coital spotting, vaginal discharge, and in advanced cases, pelvic or low back pain with sciatic nerve toot type of pain radiating down to the lower back of the lower extremities.


[0252] Women typically receive annual gynecological examination that includes PAP smears which analyses the cytological abnormalities of the cervical epithelial cells. In 1988, a National Institute of Health consensus panel was formed and had a uniformed terminology for reporting cervical cytology. Low grade squamous intraepithelial lesions (LSIL) encompassing HPV changes are considered Grade I CIN. High-grade lesions encompassing Grade II & III CIN present high risk to progress cervical cancer. If the PAP smear results are positive, the next step could be colposcopy and directed biopsy. In patients with gross evidence of tumour, diagnosis is usually confirmed by a directed cervical punch biopsy (Benedet J L et al, International Journal of Gynecology & Obstetrics, 70 (2000), 209-262).


[0253] Women diagnosed with CIN of various Grades can be treated with low-dose nitrolycerin, either topically, vaginally, or parenterally, alone or in combination with the standard therapies as recommended by the FIGO Committee on Gynecologic Oncology. Regressing on the Grades of CIN will considered as treatment success. Furthermore, if patients had a Grade II or III CIN, and fail to progress to a confirmed diagnosis of cervical cancer with nitrolycerin treatment, one can conclude that the nitroglycerin treatment has successfully prevent the progression to malignant phenotype. Patients that are in the high risk group, e.g. multiple partners with high existence or potential for HPV infection can stay on the low-dose nitroglycerin treatment to prevent recurrence.



VIII. Example 8


Prolong the Effective Treatment Period and Enhance the Overall Effectiveness of Treating Hormone Refractory/Insensitive Breast Cancer Using Fourth-Line Treatment Option by Oral PDE V Inhibitor

[0254] Breast cancer is the most common cancer and the second leading cancer death in women. In the United States, there are approximately 100,000 women alive with metastatic breast cancer. Once metastases have developed, breast cancer is rarely, if ever, a “curable” condition. When managing metastatic disease, the treatment objectives are to optimize treatment response, improve survival, and balance these goals with maintaining highest possible quality of life. Some treatments are palliative in nature. With the exception of Herceptin (specific for the HER2 oncoprotein), cytotoxic chemotherapeutic agents remains the standard for advanced hormone insensitive breast cancer.


[0255] Advanced breast cancer patients where cancer had progressed on anthracycline, taxane and capecitabine treatment can be considered for the following treatment in order to improve the therapeutic index (relief of cancer symptoms vs. the likelihood of treatement-related toxicities) and overall quality of life. While being treated with one of the fourth-line treatments (e.g., Doxil, navelbine, gemcitabine), the breast cancer patients could also take oral PDE V inhibitor, Viagra 25 mg once daily or other longer lasting PDE V inhibitor such as vanadafil. Overall increase in survival rate and or survival time of these women in comparison with the historical database of a given fourth-line treatment will be considered a treatment success of the PDEV inhibitor, Viagra. Enhancing the quality of life, reduction of pain and discomfort during the survival period is also considered as support of Viagra effectiveness in treating breast cancer patients using fourth-line treatment approach. Viagra could also be used with glucocorticoids such as prednisone to provide additional palliative relief of cancer symptoms.



IX. Example 9


Preventing the Progression of Prostate Cancer

[0256] Prostate cancer is an important public health concern, representing the most common visceral cancer and the second leading cause of cancer deaths of North American males. In 1999, the American Cancer Society estimated that approximately 179,300 new cases of prostate cancer will be diagnosed in the United States and about 37,000 men could die of the disease (Landis et al. CA Cancer J Clin 1999 49:8-31). Despite this enormous prevalence, optimal managementof both localized and metastatic disease remains elusive. Although screening efforts attempt to detect cancer at early stages, it has been estimated that 25% of men diagnosed with prostate cancer will eventually succumb to metastatic disease (Landis et al CA Cancer J Clin 1999 49:8-31).


[0257] Radical prostatectomy is considered a gold therapy treatment of localized prostate cancer and removal of all prostatic tissue should result in undetectable serum PSA within a month if all disease has been eradicated (Landis et al. CA Cancer J Clin 1999 49:8-31); therefore, longitudinal measurement of PSA is currently the most sensitive method for detecting cancer persistence, relapse and progression following radical prostatectomy (Landis et al. CA Cancer J Clin 1999 49:8-31). Except for a few anecdotal reports of prostate cancer relapse in the absence of a PSA rise, clinical evidence of local or distant cancer failure is preceded by months to years with biochemical evidence (that is, a detectable PSA; Landis et al. CA Cancer J Clin 1999 49:8-31). The ten-year actuarial incidence of biochemical failure after radical prostatectomy for clinically localized prostate cancer ranges anywhere from 27% to 57% (Zeitman et al. Urology 1994 43:828-833). A ten-year clinical local recurrence rate of 8%, clinical distant recurrence rate of 9% and any evidence of failure (biochemical or clinical) of 32% have been reported. The median time to PSA evidence of treatment failure ranges from 19 to 24 months and the median interval from biochemical recurrence to clinical evidence of disease is an additional 19 months (range: 7 to 71 months).


[0258] Men diagnosed with prostate cancer can be treated with a low-dose NO mimetic such as nitroglycerin or isosorbide dinitrate, preferably orally, sublingually, topically, or parenterally, alone or in combination with the standard therapies (e.g., anti-androgen therapy, radical prostatectomy). Exemplary low-dose NO mimetic therapies are set forth herein in Table 1. Regression or stabilization of serial PSA values is considered a treatment success. Furthermore, if subjects have prostate cancer and fail to progress to a later stage of prostate cancer with low-dose NO mimetic such as nitroglycerin or isosorbide dinitrate treatment, one can conclude that the NO mimetic treatment has successfully prevented the progression of prostate cancer. Subjects in the high-risk group, e.g., with short PSA doubling times or family history can stay on a low-dose NO mimetic (e.g., nitroglycerin or isosorbide dinitrate) treatment indefinitely to prevent development, progression or recurrence of the disease.



X. Example 10


Preventing the Progression of Breast Cancer

[0259] There is evidence that the local tumor microenvironment plays an important role in determining the behavior of the tumor cells. Hypoxia (pO2 values of less than 10 mmHg) within the solid tumor mass is an independent marker of a poor clinical outcome for patients with a variety of cancers, and in particular breast cancer. The risk of mortality is significantly increased if a primary tumor undergoes metastasis to a distant site within the body.


[0260] Females diagnosed with breast cancer can be treated with a low-dose NO mimetic such as nitroglycerin or isosorbide dinitrate, preferably orally, sublingually, topically, or parenterally, alone or in combination with the standard chemotherapies (e.g. taxol). Exemplary low-dose NO mimetic therapies are set forth herein in Table 1. Regression or stabilization of a primary tumor is considered a treatment success. Subjects in the high-risk group (e.g. family history) can stay on a low-dose NO mimetic (e.g. nitroglycerin or isosorbide dinitrate) treatment indefinitely to prevent development, progression or recurrence of the disease.



XI. Example 11


Effect of GTN Therapy on PSA Levels

[0261] PSA levels, a biochemical marker of prostate cancer recurrence, were determined following GTN administration in patients with prostate cancer wherein the prostate is still intact.


[0262] In this prostate cancer patient, he received radiation therapy in 1991 as the treatment choice. Subsequently, he was place on anti-androgen therapy as the next line of treatment. One year later, there was an increase of serum PSA level as an indicator of cancer relapse. As shown in FIG. 16, the rate of increase in circulating PSA levels decreased in this patient following commencement of two separate episodes of administration of GTN at 0.03 mg/hour, 24 hours a day, for approximately one month each. In both periods, a reduction of PSA velocity was observed with GTN treatment. Following the commencement of a third episode of administration of GTN at 0.03 mg/hour, 24 hours a day, for approximately one month, a decrease in PSA levels was again observed. This result suggest that NO mimetics such as GTN could improve the effectiveness of anti-androgen therapy treatment alone in 1) time to biochemical failure indicated by continuous increase in PSA velocity and 2) in prolonging the time to full relapse of prostate cancer in men.


[0263] Further, as shown in FIG. 17, chronic administration of GTN, transdermally, at a concentration of 0.03 mg/hour dramatically improved the efficacy, as determined by circulating PSA levels, of radiation therapy. Two months after chronic GTN therapy was begun, this patient was administered radiation therapy. As shown in FIG. 17, this combination therapy accelerated the rate of PSA decrease to within three months. The expected average for a similar decrease in PSA levels following radiation therapy alone is twelve months. This result support the use of NO mimetics in enhancing the effectiveness of radiation therapy in treating prostate cancer patients by 1) decrease the serum marker of prostate cancer, i.e. PSA level, 2) potential lead to remission of prostate cancer and 3) maintaining prostate cancer at dormant stage indefinitely.



XII. Example 12

[0264] Prostate cancer is now the most commonly diagnosed cancer and the second most common cause of cancer death in men. Prostate cancer is mostly adenocarcinoma that arise from high-grade prostate intraepithelial neoplasia (PIN) which is present in 4-16.5% of needle biopsies and is strongly predictive of co-existing carcinoma, thus warranting a repeat biopsy. Early prostate cancer patients can be effectively treated with available androgen suppression therapy.


[0265] The treatment of hormone refractory advanced prostate cancer remains a clinical challenge. In hormone-refractory/insensitive prostate cancer (HRPC) patients who failed one non-estramustine, taxane-containing regimen and entering an estramustine treatment regimen (e.g. 14 mg/kg/day PO in 3-4 individual doses) will be provided with topical nitroglycerin ointment. The nitroglycerin ointment will be applied 4-5 times daily to provide sustained plasma level of nitric oxide. While under the nitroglycerin and estramustine treatment, the PSA levels will be measured monthly. The effect of nitroglycerin treatment will be measured against historical database of estramustine users. A statistic of ≧40% patients with ≧50% decrease in PSA levels or velocity will be considered a nitroglycerin treatment success. The overall increase in the expected median survival time of 5 months of estramustine mono-therapy, the reduction of pain score and the use of pain medication, or improvement in patients' ability to maintain cognitive function and to copy with the advanced disease will also considered as clinically meaningful to add nitroglycerin treatment to the standard of care of these patients.



Example 13


Preventing the Recurrence of Malignant Breast Cancer Following Lumpectomy Procedure

[0266] Patients with localized early stage non-metastatic breast cancer may elected to undergo a lumpectomy procedure to remove the malignant tissue. Prior to the surgery, patients may start with first line anti-malignant therapy to reduce the tumor volume. To improve the effectiveness of this treatment, patients could start taking nitric oxide mimetics such as Viagra (25 mg or 50 mg) daily continuously during the first line therapy, right after the surgery and chronically post-surgical procedure to prevent relapse of the breast cancer or other form of cancer. Prolong remission period against the history database and/or better quality of life will be considered a treatment success.


[0267] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.


Claims
  • 1. A method for controlling, treating and/or preventing cancer, malignancies, neoplasm, hyperplasia, hypertrophy, dysplasia and/or tumor angiogenesis, said method comprising: administering a nitric oxide mimetic to an animal thereof, to control, treat and/or prevent said cancer, malignancies, neoplasm, hyperplasia, hypertrophy, dysplasia and/or tumor angiogenesis.
  • 2. The method according to claim 1, wherein said nitric oxide mimetic is administered at a low dose.
  • 3. The method according to claim 1, wherein said nitric oxide mimetic is administered at a level which delays and/or reduces development of tolerance to the nitric oxide mimetic and/or unwanted side effects, including headache, flushing and hypotension.
  • 4. The method according to claim 1, wherein said nitric oxide mimetic is administered alone or in combination with an antimalignant therapeutic agent.
  • 5. The method according to claim 1, wherein said nitric oxide mimetic; (1) inhibits the metastatic potential of a tumor or malignant cell phenotype preferably by decreasing the invasiveness, progression, growth and/or metastases of cells exhibiting a malignant phenotype; inhibiting the survival and/or growth of cells exhibiting a malignant phenotype; decreasing the progression and/or metastases of cells exhibiting a malignant phenotype; increasing the regression of cells exhibiting a malignant phenotype; and/or facilitating the killing of cells exhibiting a malignant phenotype; (2) maintains a malignant tumor in a dormant or quiescent state at its primary and/or secondary site; (3) enhances the efficacy of, and/or prevents or decreases the resistance to an antimalignant therapeutic modality; or (4) inhibits or prevents tumor angiogenesis in animals at high risk of developing cancer and/or exposed to factors known to decrease nitric oxide activity in an animal, optionally wherein said factors include decreased arginine levels, exposure to nitric oxide synthase antagonists, exposure to nitric oxide scavengers, changes in nitric oxide synthase expression, change in cofactors, glucose deprivation, surgical procedures, administration of anaesthetic agents, administration of pharmacologic agents which alter circulation, traumatic injuries, physical trauma, blood loss, decreased blood volume, or hemorrhage, or combinations thereof.
  • 6. The method according to claim 1, wherein the cells exhibiting the malignancies are selected from malignant cells, invasive cells, cells and tissue[s] that facilitate the malignant process, and combinations thereof, optionally wherein the malignant cell phenotype is controlled, treated or prevented by improving response to an antimalignant therapeutic modality.
  • 7. The method according to claim 1, wherein cancer is diagnosed or monitored by measuring a tumor selective marker present in said animal.
  • 8. The method according to claim 7, wherein said nitric oxide mimetic decreases or decelerates increases of the level of said tumor marker.
  • 9. The method according to claim 1, wherein said cancer comprises gastric cancer, gastrointestinal cancer, testicular cancer, prostate cancer, prostatic adenocarcinoma, breast cancer, metastatic melanoma, or lung cancer, or combinations thereof; optionally wherein the cancer or other malignancies, neoplasm, hyperplasia, hypertrophy, dysplasia and/or tumor angiogenesis in an animal comprises benign prostatic hyperplasia or molar pregnancy.
  • 10. The method according to claim 1, wherein said nitric oxide mimetic comprises nitric oxide, a nitric oxide donor, a compound that generates or releases nitric oxide through biotransformation, a compound that generates nitric oxide spontaneously or spontaneously releases nitric oxide, or a compound which generates nitric oxide, or combinations thereof.
  • 11. The method according to claim 10, wherein said nitric oxide mimetic is (1) a nitric oxide donor selected from nitroglycerin (GTN), isosorbide 5-mononitrate (ISMN), isosorbide dinitrate (ISDN), pentaerythritol tetranitrate (PETN), erthrityl tetranitrate (ETN), N-hydroxyl-L-arginine (NOHA), N6-(1-iminoethyl)lysine) (L-NIL), L-N5-(1-iminoethyl) ornithine (LN-NIO), Nw-methyl-L-arginine (L-NMMA), S-nitrosogluthathione (SNOG), S,S-dinitrosodithiol (SSDD), [N-[2(nitroxyethyl)]-3-pyridinecarboxamide (nicorandil), sodium nitroprusside (SNP), S-nitroso-N-acetylpenicilamine (SNAP), 3-morpholino-sydnonimine (SIN-1), molsidomine, DEA-NONOate (2(N,N-diethylamino)-diazenolate-2-oxide), and spermine NONOate (N-[4-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]butyl-1,3-propanediamine; (2) a compound that activates stages of NO pathway, a compound which enables or facilitates NO utilization by a cell, a compound which directly activates guanylyl cyclase, or a phosphodiesterase inhibitor, or combinations thereof, (3) a type I, II, III, IV or V phosphodiesterase inhibitor, or combinations thereof; or (4) a protein kinase G activator.
  • 12. The method according to claim 4, wherein said antimalignant therapeutic agent includes radiation therapy, thermal therapy, immunotherapy, or chemotherapy, or combinations thereof, optionally wherein the antimalignant therapeutic modality comprises radiation therapy, and said nitric oxide mimetic is a nitric oxide, a nitric oxide donor, a compound that generates or releases nitric oxide through biotransformation, or a compound that generates nitric oxide spontaneously or spontaneously releases nitric oxide only in the presence of oxygen, or combinations thereof, wherein said nitric oxide mimetic is administered during the radiation therapy.
  • 13. The method according to claim 12, wherein said chemotherapy comprises administration of chemotherapeutic agent that is an anti-angiogenic agent, an antimetabolite, an antibiotic, an endothelin activating agent, an enzyme inhibitor, a hormonal agent, ocreotide acetate, a microtubule-disruptor agent, a microtubule-stabilizing agent, a vinca alkaloid, a epipodophyllotoxin, a toposiomerase inhibitor; a prenyl-protein transferase inhibitor, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, a platinum coordination complex, a biological response modifier, a growth factor, an immune modulator, or a monoclonal antibody, or a combination thereof.
  • 14. The method according to claim 1, wherein the dose of nitric oxide mimetic is at least 3- to 10,000-fold lower, preferably 100-10,000 fold lower than a dose of nitric oxide mimetic that produces vasodilation.
  • 15. The method according to claim 1, wherein said nitric oxide mimetic is a known vasodilatory compound and said mimetic is administered at a dose of at least 3- to 10,000-fold lower, preferably 100-10,000 fold lower than the dose of nitric oxide mimetic known to produce vasodilation.
  • 16. The method according to claim 1, wherein said nitric oxide mimetic is selected from the group consisting of a calcium channel blocker, an α-adrenergic receptor antagonist, a β-adrenergic receptor agonist, a phosphodiesterase inhibitor, a cAMP-dependent protein kinase activator, a superoxide scavenger, a potassium channel activator, a benzodiazepine, an adrenergic nerve inhibitor, an antidiarrheal agent, a HMG-CoA reductase inhibitor, an adenosine receptor modulator, a adenylyl cyclase activator, an endothelin receptor antagonist, a bisphosphonate, a cGMP-dependent protein kinase activator, a guanylyl cyclase activator and a SOC inhibitor.
CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. Provisional Application Serial Nos. 60/362,969 filed Mar. 6, 2002, and 60/362,620 filed Mar. 7, 2002, and U.S. patent application Ser. No. 10/042,039, filed Oct. 25, 2001, which are herein incorporated by reference. U.S. patent application Ser. No. 10/042,039 is a continuation-in-part of U.S. application Ser. No. 09/842,547, filed Apr. 26, 2001, which claims the benefit of U.S. Provisional Application Nos. 60/277,469, filed Mar. 21, 2001, and 60/199,757, filed Apr. 26, 2000, which are herein incorporated by reference in their entirety.

Provisional Applications (4)
Number Date Country
60362620 Mar 2002 US
60362969 Mar 2002 US
60277469 Mar 2001 US
60199757 Apr 2000 US
Continuation in Parts (2)
Number Date Country
Parent 10042039 Oct 2001 US
Child 10384499 Mar 2003 US
Parent 09842547 Apr 2001 US
Child 10384499 Mar 2003 US