AU2008356891A1 - Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin - Google Patents

Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin Download PDF

Info

Publication number
AU2008356891A1
AU2008356891A1 AU2008356891A AU2008356891A AU2008356891A1 AU 2008356891 A1 AU2008356891 A1 AU 2008356891A1 AU 2008356891 A AU2008356891 A AU 2008356891A AU 2008356891 A AU2008356891 A AU 2008356891A AU 2008356891 A1 AU2008356891 A1 AU 2008356891A1
Authority
AU
Australia
Prior art keywords
cancer
inositol
tripyrophosphate
myo
composition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2008356891A
Inventor
Konstantina C. Fylaktakidou
Ruth Greferath
Jean-Marie Lehn
Claude Nicolau
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universite de Strasbourg
NormOxys Inc
Original Assignee
Universite de Strasbourg
NormOxys Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universite de Strasbourg, NormOxys Inc filed Critical Universite de Strasbourg
Publication of AU2008356891A1 publication Critical patent/AU2008356891A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/093Polyol derivatives esterified at least twice by phosphoric acid groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/6574Esters of oxyacids of phosphorus
    • C07F9/65746Esters of oxyacids of phosphorus the molecule containing more than one cyclic phosphorus atom

Description

WO 2009/145751 PCT/US2008/006837 CALCIUM/SODIUM SALT OF INOSITOL TRIPYROPHOSPHATE AS AN ALLOSTERIC EFFECTOR OF HEMOGLOBIN 5 FIELD OF THE INVENTION The present invention is directed to compositions and methods for using the mixed calcium/sodium salt of inositol-tripyrophosphate (ITPP-Ca/Na) to enhance oxygen delivery by red blood cells. ITPP-Ca/Na is an allosteric effector of hemoglobin which has the ability to cross the plasma membrane of red blood cells and 10 lower the oxygen affinity of the hemoglobin of red blood cells. The present invention is further directed to the use of ITPP-Ca/Na to inhibit angiogenesis and enhance radiation sensitivity of hypoxic tumors. The present invention is further directed to the use of ITPP-Ca/Na to enhance the partial pressure of oxygen (P0 2 ) in hypoxic tumors. 15 BACKGROUND OF THE INVENTION In the vascular system of an adult human being, blood has a volume of about 5 to 6 liters. Approximately one half of this volume is occupied by cells, including red blood cells (erythrocytes), white blood cells (leukocytes), and blood 20 platelets. Red blood cells comprise the majority of the cellular components of blood. Plasma, the liquid portion of blood, is approximately 90 percent water and 10 percent various solutes. These solutes include plasma proteins, organic metabolites and waste products, and inorganic compounds. The major function of red blood cells is to transport oxygen from the 25 lungs to the tissues of the body, and transport carbon dioxide from the tissues to the lungs for removal. Very little oxygen is transported by the blood plasma because oxygen is only sparingly soluble in aqueous solutions. Most of the oxygen carried by the blood is transported by the hemoglobin of the erythrocytes. Erythrocytes in mammals do not contain nuclei, mitochondria or any other intracellular organelles, 30 and they do not use oxygen in their own metabolism. Red blood cells contain about 35 percent by weight hemoglobin, which is responsible for binding and transporting oxygen.
WO 2009/145751 PCT/US2008/006837 Hemoglobin is a protein having a molecular weight of approximately 64,500 daltons. It contains four polypeptide chains and four heme prosthetic groups in which iron atoms are bound in the ferrous state. Normal globin, the protein portion of the hemoglobin molecule, consists of two alpha chains and two beta chains. Each 5 of the four chains has a characteristic tertiary structure in which the chain is folded. The four polypeptide chains fit together in an approximately tetrahedral arrangement, to constitute the characteristic quaternary structure of hemoglobin. There is one heme group bound to each polypeptide chain which can reversibly bind one molecule of molecular oxygen. When hemoglobin combines with oxygen, oxyhemoglobin is 10 formed. When oxygen is released, the oxyhemoglobin is reduced to deoxyhemoglobin. Delivery of oxygen to tissues, including tumors, depends upon a number of factors including, but not limited to, the volume of blood flow, the number of red blood cells, the concentration of hemoglobin in the red blood cells, the oxygen 15 affinity of the hemoglobin and, in certain species, on the molar ratio of intraerythrocytic hemoglobins with high and low oxygen affinity. The oxygen affinity of hemoglobin depends on four factors as well, namely: (1) the partial pressure of oxygen; (2) the pH; (3) the concentration of 2,3-diphosphoglycerate (DPG) in the hemoglobin; and (4) the concentration of carbon dioxide. In the lungs, at an oxygen 20 partial pressure of 100 mm Hg, approximately 98% of circulating hemoglobin is saturated with oxygen. This represents the total oxygen transport capacity of the blood. When fully oxygenated, 100 ml of whole mammalian blood can carry about 21 ml of gaseous oxygen. The effect of the partial pressure of oxygen and the pH on the ability of 25 hemoglobin to bind oxygen is best illustrated by examination of the oxygen saturation curve of hemoglobin. An oxygen saturation curve plots the percentage of total oxygen-binding sites of a hemoglobin molecule that are occupied by oxygen molecules when solutions of the hemoglobin molecule are in equilibrium with different partial pressures of oxygen in the gas phase. 30 The oxygen saturation curve for hemoglobin is sigmoid. Thus, binding the first molecule of oxygen increases the affinity of the remaining hemoglobin for binding additional oxygen molecules. As the partial pressure of oxygen is increased, -2- WO 2009/145751 PCT/US2008/006837 a plateau is approached at which each of the hemoglobin molecules is saturated and contains the upper limit of four molecules of oxygen. The reversible binding of oxygen by hemoglobin is accompanied by the release of protons, according to the equation: 5 HHb+ +02 = HbO 2 + H* Thus, an increase in the pH will pull the equilibrium to the right and cause hemoglobin to bind more oxygen at a given partial pressure. A decrease in the 10 pH will decrease the amount of oxygen bound. In the lungs, the partial pressure of oxygen in the air spaces is approximately 90 to 100 mm Hg and the pH is also high relative to normal blood pH (up to 7.6). Therefore, hemoglobin will tend to become almost maximally saturated with oxygen in the lungs. At that pressure and pH, hemoglobin is approximately 15 98 percent saturated with oxygen. On the other hand, in the capillaries in the interior of the peripheral tissues, the partial pressure of oxygen is only about 25 to 40 mm Hg and the pH is also nearly neutral (about 7.2 to 7.3). Because muscle cells use oxygen at a high rate, thereby lowering the local concentration of oxygen, the release of some of the bound oxygen to the tissue is favored. As the blood passes through the 20 capillaries in the muscles, oxygen will be released from the nearly saturated hemoglobin in the red blood cells into the blood plasma and then into the muscle cells. Hemoglobin will release about a fourth of its bound oxygen as it passes through the muscle capillaries, so that when it leaves the muscle, it will be only about 75 percent saturated. In general, the hemoglobin in the venous blood leaving the tissue cycles 25 between about 65 and 97 percent saturation with oxygen in its repeated circuits between the lungs and the peripheral tissues. Thus, oxygen partial pressure and pH function together to effect the release of oxygen by hemoglobin. A third important factor in regulating the degree of oxygenation of hemoglobin is the allosteric effector 2,3-diphosphoglycerate (DPG). DPG is the 30 normal physiological effector of hemoglobin in mammalian erythrocytes. DPG regulates the oxygen-binding affinity of hemoglobin in the red blood cells in relationship to the oxygen partial pressure in the lungs. The higher the concentration of DPG in the cell, the lower the affinity of hemoglobin for oxygen. -3 - WO 2009/145751 PCT/US2008/006837 When the delivery of oxygen to the tissues is chronically reduced, the concentration of DPG in the erythrocytes is higher than in normal individuals. For example, at high altitudes the partial pressure of oxygen is significantly less. Correspondingly, the partial pressure of oxygen in the tissues is less. Within a few 5 hours after a normal human subject moves to a higher altitude, the DPG level in the red blood cells increases, causing more DPG to be bound and the oxygen affinity of the hemoglobin to decrease. Increases in the DPG level of red cells also occur in patients suffering from hypoxia. This adjustment allows the hemoglobin to release its bound oxygen more readily to the tissues to compensate for the decreased oxygenation 10 of hemoglobin in the lungs. The reverse change occurs when people are acclimated to high altitudes and descend to lower altitudes. As normally isolated from blood, hemoglobin contains a considerable amount of DPG. When hemoglobin is "stripped" of its DPG, it shows a much higher affinity for oxygen. When DPG is increased, the oxygen binding affinity of 15 hemoglobin decreases. A physiologic allosteric effector such as DPG is therefore essential for the normal release of oxygen from hemoglobin in the tissues. While DPG is the normal physiologic effector of hemoglobin in mammalian red blood cells, phosphorylated inositols are found to play the same role in the erythrocytes of some birds and reptiles. Although inositol hexaphosphate (IHP) 20 is unable to pass through the mammalian erythrocyte membrane, it is capable of combining with hemoglobin of mammalian red blood cells at the binding site of DPG to modify the allosteric conformation of hemoglobin, the effect of which is to reduce the affinity of hemoglobin for oxygen. For example, DPG can be replaced by IHP, which is far more potent than DPG in reducing the oxygen affinity of hemoglobin. 25 IHP has a 1000-fold higher affinity to hemoglobin than DPG (R. E. Benesch et al., Biochemistry, Vol. 16, pages 2594-2597 (1977)) and increases the P 5 o of hemoglobin up to values of 96.4 mm, Hg at pH 7.4, and 37 degrees C. (J. Biol. Chem., Vol. 250, pages 7093-7098 (1975)). The oxygen release capacity of mammalian red blood cells can be 30 enhanced by introducing certain allosteric effectors of hemoglobin into erythrocytes, thereby decreasing the affinity of hemoglobin for oxygen and improving the oxygen economy of the blood. This phenomenon suggests various medical applications for -4- WO 2009/145751 PCT/US2008/006837 treating individuals who are experiencing lowered oxygenation of their tissues due to the inadequate function of their lungs or circulatory system. Because of the potential medical benefits to be achieved from the use of these modified erythrocytes, various techniques have been developed in the prior 5 art to enable the encapsulation of allosteric effectors of hemoglobin in erythrocytes. Accordingly, numerous devices have been designed to assist or simplify the encapsulation procedure. The encapsulation methods known in the art include osmotic pulse (swelling) and reconstitution of cells, controlled lysis and resealing, incorporation of liposomes, and electroporation. Current methods of electroporation 10 make the procedure commercially impractical on a scale suitable for commercial use. The following references describe the incorporation of polyphosphates into red blood cells by the interaction of liposomes loaded with IHP: Gersonde, et al., "Modification of the Oxygen Affinity of Intracellular Hemoglobin by Incorporation of Polyphosphates into Intact Red Blood Cells and Enhanced 02 Release in the Capillary 15 System", Biblthca. Haemat., No. 46, pp. 81-92 (1980); Gersonde, et al., "Enhancement of the 02 Release Capacity and of the Bohr-Effect of Human Red Blood Cells after Incorporation of Inositol Hexaphosphate by Fusion with Effector-Containing Lipid Vesicles", Origins of Cooperative Binding of Hemoglobin (1982); and Weiner, "Right Shifting of Hb-0 2 Dissociation in Viable Red Cells by Liposomal Technique," 20 Biology of the Cell, Vol. 47, (1983). Additionally, U.S. Pat. Nos. 4,192,869, 4,321,259, and 4,473,563 to Nicolau et al. describe a method whereby fluid-charged lipid vesicles are fused with erythrocyte membranes, depositing their contents into the red blood cells. In this manner, it is possible to transport allosteric effectors, such as IHP into erythrocytes, 25 where due to its much higher binding constant IHP replaces DPG at its binding site in hemoglobin. In accordance with the liposome technique, IHP is dissolved in a phosphate buffer until the solution is saturated and a mixture of lipid vesicles is suspended in the solution. The suspension is then subjected to ultrasonic treatment or 30 an injection process, and then centrifuged. The upper suspension contains small lipid vesicles containing IHP, which are then collected. Erythrocytes are added to the collected suspension and incubated, during which time the lipid vesicles containing IHP fuse with the cell membranes of the erythrocytes, thereby depositing their -5- WO 2009/145751 PCT/US2008/006837 contents into the interior of the erythrocyte. The modified erythrocytes are then washed and added to plasma to complete the product. The drawbacks associated with the liposomal technique include poor reproducibility of the IHP concentrations incorporated in the red blood cells and 5 significant hemolysis of the red blood cells following treatment. Additionally, commercialization is not practical because the procedure is tedious and complicated. In an attempt to solve the drawbacks associated with the liposomal technique, a method of lysing and the resealing red blood cells was developed. This method is described in the following publication: Nicolau, et al., "Incorporation of 10 Allosteric Effectors of Hemoglobin in Red Blood Cells. Physiologic Effects," Biblthca. Haemat., No. 51, pp. 92-107, (1985). Related U.S. Pat. Nos. 4,752,586 and 4,652,449 to Ropars et al. also describe a procedure of encapsulating substances having biological activity in human or animal erythrocytes by controlled lysis and resealing of the erythrocytes, which avoids the red blood cell-liposome interactions. 15 The technique is best characterized as a continuous flow dialysis system, which functions in a manner similar to the osmotic pulse technique. Specifically, the primary compartment of at least one dialysis element is continuously supplied with an aqueous suspension of erythrocytes, while the secondary compartment of the dialysis element contains an aqueous solution which is hypotonic 20 with respect to the erythrocyte suspension. The hypotonic solution causes the erythrocytes to lyse. The erythrocyte lysate is then contacted with the biologically active substance to be incorporated into the erythrocyte. To reseal the membranes of the erythrocytes, the osmotic and/or oncotic pressure of the erythrocyte lysate is increased and the suspension of resealed erythrocytes is recovered. 25 In related U.S. Pat. Nos. 4,874,690 and 5,043,261 to Goodrich et al., a related technique involving lyophilization and reconstitution of red blood cells is disclosed. As part of the process of reconstituting the red blood cells, the addition of various polyanions, including IHP, is described. Treatment of the red blood cells according to the process disclosed results in a cell with unaffected activity. 30 Presumably, the IHP is incorporated into the cell during the reconstitution process, thereby maintaining the activity of the hemoglobin. -6- WO 2009/145751 PCT/US2008/006837 In U.S. Pat. Nos. 4,478,824 and 4,931,276 to Franco et al., a second related method and apparatus is described for introducing effectively non-ionic agents, including IHP, into mammalian red blood cells by effectively lysing and resealing the cells. The procedure is described as the "osmotic pulse technique." In 5 practicing the osmotic pulse technique, a supply of packed red blood cells is suspended and incubated in a solution containing a compound which readily diffuses into and out of the cells, the concentration of the compound being sufficient to cause diffusion thereof into the cells so that the contents of the cells become hypertonic. Next, a trans-membrane ionic gradient is created by diluting the solution containing 10 the hypertonic cells with an essentially isotonic aqueous medium in the presence of at least one desired agent to be introduced, thereby causing diffusion of water into the cells with a consequent swelling and an increase in permeability of the outer membranes of the cells. This "osmotic pulse" causes the diffusion of water into the cells and a resultant swelling of the cells which increase the permeability of the outer 15 cell membrane to the desired agent. The increase in permeability of the membrane is maintained for a period of time sufficient only to permit transport of at least one agent into the cells and diffusion of the compound out of the cells. Polyanions which may be used in practicing the osmotic pulse technique include pyrophosphate, tripolyphosphate, phosphorylated inositols, 2,3 20 diphosphoglycerate (DPG), adenosine triphosphate, heparin, and polycarboxylic acids which are water-soluble, and non-disruptive to the lipid outer bilayer membranes of red blood cells. The osmotic pulse technique has several shortcomings including low yield of encapsulation, incomplete resealing, loss of cell content and a corresponding 25 decrease in the life span of the cells. The technique is tedious, complicated and unsuited to automation. For these reasons, the osmotic pulse technique has had little commercial success. Another method for encapsulating various biologically-active substances in erythrocytes is electroporation. Electroporation has been used for 30 encapsulation of foreign molecules in different cell types, including IHP in red blood cells, as described in Mouneimne, et al., "Stable rightward shifts of the oxyhemoglobin dissociation curve induced by encapsulation of inositol hexaphosphate -7- WO 2009/145751 PCT/US2008/006837 in red blood cells using electroporation," FEBS, Vol. 275, No. 1, 2, pp. 117-120 (1990). Also, see U.S. Patent No. 5,612,207. Angiogenesis is the generation of new blood vessels into a tissue or organ and is related to oxygen tension in the tissues. Under normal physiological 5 conditions, humans and animals undergo angiogenesis only in very specific, restricted situations. For example, angiogenesis is normally observed in wound healing, fetal and embryonal development, and formation of the corpus luteum, endometrium and placenta. Angiogenesis is controlled through a highly regulated system of 10 angiogenic stimulators and inhibitors. The control of angiogenesis is altered in certain disease states and, in many cases, pathological damage associated with the diseases is related to uncontrolled angiogenesis. Both controlled and uncontrolled angiogenesis are thought to proceed in a similar manner. Endothelial cells and pericytes, surrounded by a basement membrane, form capillary blood vessels. 15 Angiogenesis begins with the erosion of the basement membrane by enzymes released by endothelial cells and leukocytes. Endothelial cells, lining the lumen of blood vessels, then protrude through the basement membrane. Angiogenic stimulants induce the endothelial cells to migrate through the eroded basement membrane. The migrating cells form a "sprout" off the parent blood vessel where the endothelial cells 20 undergo mitosis and proliferate. The endothelial sprouts merge with each other to form capillary loops, creating a new blood vessel. Persistent, unregulated angiogenesis occurs in many disease states, tumor metastases, and abnormal growth by endothelial cells. The diverse pathological disease states in which unregulated angiogenesis is present have been grouped 25 together as angiogenic-dependent or angiogenic-associated diseases. The hypothesis that tumor growth is angiogenesis-dependent was first proposed in 1971. (Folkman, New Eng. J. Med., 285:1182-86 (1971)). In its simplest terms, this hypothesis states: "Once tumor 'take' has occurred, every increase in tumor cell population must be preceded by an increase in new capillaries converging 30 on the tumor." Tumor 'take' is currently understood to indicate a prevascular phase of tumor growth in which a population of tumor cells occupying a few cubic millimeters volume, and not exceeding a few million cells, can survive on existing host microvessels. Expansion of tumor volume beyond this phase requires the -8- WO 2009/145751 PCT/US2008/006837 induction of new capillary blood vessels. For example, pulmonary micrometastases in the early prevascular phase in mice would be undetectable except by high power microscopy on histological sections. Angiogenesis has been associated with a number of different types of 5 cancer, including solid tumors and blood-borne tumors. Solid tumors with which angiogenesis has been associated include, but are not limited to, rhabdomyosarcomas, retinoblastoma, Ewing's sarcoma, neuroblastoma, and osteosarcoma. Angiogenesis is also associated with blood-borne tumors, such as leukemias, any of various acute or chronic neoplastic diseases of the bone marrow in which unrestrained proliferation of 10 white blood cells occurs, usually accompanied by anemia, impaired blood clotting, and enlargement of the lymph nodes, liver and spleen. It is believed that angiogenesis plays a role in the abnormalities in the bone marrow that give rise to leukemia tumors and multiple myeloma diseases. One of the most frequent angiogenic diseases of childhood is the 15 hemangioma. A hemangioma is a tumor composed of newly formed blood vessels. In most cases, the tumors are benign and regress without intervention. In more severe cases, the tumors progress to large cavernous and infiltrative forms and create clinical complications. Systemic forms of hemangiomas, hemangiomatoses, have a high mortality rate. Therapy-resistant hemangiomas exist that cannot be treated with 20 therapeutics currently in use. Another angiogenesis associated disease is rheumatoid arthritis. The blood vessels in the synovial lining of the joints undergo angiogenesis. In addition to forming new vascular networks, the endothelial cells release factors and reactive oxygen species that lead to pannus growth and cartilage destruction. Angiogenesis 25 may also play a role in osteoarthritis. The activation of the chondrocytes by angiogenic-related factors contributes to the destruction of the joint. At a later stage, the angiogenic factors promote new bone growth. Therapeutic intervention that prevents the cartilage destruction could halt the progress of the disease and provide relief for persons suffering with arthritis. 30 Chronic inflammation may also involve pathological angiogenesis. Such diseases as ulcerative colitis and Crohn's disease show histological changes with the ingrowth of new blood vessels into inflamed tissues. Bartonelosis, a bacterial infection found in South America, can result in a chronic stage that is characterized by -9- WO 2009/145751 PCT/US2008/006837 proliferation of vascular endothelial cells. Another pathological role associated with angiogenesis is found in atherosclerosis. The plaques formed within the lumen of blood vessels have been shown to have angiogenic stimulatory activity. As mentioned above, several lines of evidence indicate that 5 angiogenesis is essential for the growth and persistence of solid tumors and their metastases. Once angiogenesis is stimulated, tumors upregulate the production of a variety of angiogenic factors, including fibroblast growth factors (aFGF and bFGF) and vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) [2,3]. 10 The role of VEGF in the regulation of angiogenesis has been the object of intense investigation [5-10]. Whereas VEGF represents a critical, rate-limiting step in physiological angiogenesis, it appears to be also important in pathological angiogenesis, such as that associated with tumor growth [11]. VEGF is also known as vascular permeability factor, based on its ability to induce vascular leakage [13]. 15 Several solid tumors produce ample amounts of VEGF, which stimulates proliferation and migration of endothelial cells, thereby inducing neovascularization [12,13]. VEGF expression has been shown to significantly affect the prognosis of different kinds of human cancer. Oxygen tension in the tumor has a key role in regulating the expression of VEGF gene. VEGF mRNA expression is induced by exposure to low 20 oxygen tension under a variety of pathophysiological circumstances [13]. Growing tumors are characterized by hypoxia, which induces expression of VEGF and may also be a predictive factor for the occurrence of metastatic disease. What is needed, therefore, is a substantially non-toxic composition and method that can regulate oxygen tension in the tissue, especially a tumor. In addition, 25 what is needed is a simple and easily administered, preferably orally, composition that is capable of causing significant right shifts of the P 50 value for red blood cells. SUMMARY OF THE INVENTION The present invention provides a composition comprising the mixed 30 calcium/sodium salt of inositol-tripyrophosphate (ITPP-Ca/Na) that is effective in treating diseases characterized by abnormal angiogenesis. The compositions and methods of the present invention have a distinct advantage over the prior art in that -10- WO 2009/145751 PCT/US2008/006837 the compositions and methods of the present invention are substantially non-toxic and demonstrate improved solubility when compared to compositions in the prior art. The present invention also comprises a pharmaceutical composition comprising the calcium/sodium salt of ITPP and a pharmaceutically acceptable 5 adjuvant, diluent, carrier, or excipient thereof. In this pharmaceutical composition, the inositol tripyrophosphate is optimally myo-inositol 1,6:2,3:4,5 tripyrophosphate. In an alternate embodiment the composition may comprise the monocalcium tetrasodium salt of myo-inositol 1,6:2,3:4,5 tripyrophosphate. The present invention also provides for substantially non-toxic methods 10 of using the above compositions of ITPP-Ca/Na for increasing the regulated delivery of oxygen to tissues including tumors. For example, the regulation of vascular endothelial growth factor (VEGF) in a human or animal can be effected using ITPP Ca/Na which has entered the red blood cell, thus lowering the affinity for oxygen of circulating erythrocytes. In an embodiment of the present invention, ITPP-Ca/Na can 15 affect VEGF mRNA expression, protein concentration, and tumor cell proliferation. Also, a method of regulating VEGF expression, both in vitro and in vivo, using ITPP Ca/Na is contemplated and therefore within the scope of the present invention. The present invention further comprises methods for using the above compositions of ITPP-Ca/Na in pure hemoglobin and in red blood cells to deliver 20 oxygen to solid tumors, to inhibit angiogenesis and to enhance radiation sensitivity of hypoxic tumors. The present invention is further directed to the use of ITPP-Ca/Na to enhance P0 2 in hypoxic tumors. ITPP-Ca/Na is an allosteric effector of hemoglobin and is capable of reducing hemoglobin's affinity for oxygen, which enhances the release of oxygen by hemoglobin. Upon cellular demand, ITPP-Ca/Na can inhibit 25 VEGF expression in tumor cells and, thus, angiogenesis. A disease characterized by undesirable angiogenesis or undesirable angiogenesis, as defined herein includes, but is not limited to, excessive or abnormal stimulation of endothelial cells (e.g. atherosclerosis), blood borne tumors, solid tumors and tumor metastasis, benign tumors, for example, hemangiomas, acoustic 30 neuromas, neurofibromas, trachomas, and pyogenic granulomas, vascular malfunctions, abnormal wound healing, inflammatory and immune disorders, Bechet's disease, gout, or gouty arthritis, diabetic retinopathy and other ocular angiogenic diseases such as retinopathy of prematurity (retrolental fibroplasic), macular -11- WO 2009/145751 PCT/US2008/006837 degeneration, corneal graft rejection, neovascular glaucoma and Osler Weber syndrome (Osler-Weber-Rendu disease). Cancers that can be treated by the present invention include, but is not limited to, breast cancer, prostrate cancer, renal cell cancer, brain cancer, ovarian cancer, colon cancer, bladder cancer, pancreatic cancer, 5 stomach cancer, esophageal cancer, cutaneous melanoma, liver cancer, lung cancer, testicular cancer, kidney cancer, bladder cancer, cervical cancer, lymphoma, parathyroid cancer, penile cancer, rectal cancer, small intestine cancer, thyroid cancer, uterine cancer, Hodgkin's lymphoma, lip and oral cancer, skin cancer, leukemia or multiple myeloma. 10 An object of the invention is to provide a substantially non-toxic composition and method for treating cancer and other angiogenic disease states and conditions using ITPP-Ca/Na in an effective dose. Another object of the invention is to provide a composition and method for enhancing oxygen delivery to hypoxic tumors using ITPP-Ca/Na in an effective 15 dose. Yet another object of the invention is to provide a composition and method for inhibiting angiogenesis using ITPP-Ca/Na in an effective dose. A further object of the invention is to provide a composition and method for enhancing radiation sensitivity of hypoxic tumors using ITPP-Ca/Na in an 20 effective dose. It is yet another object of the invention to provide a composition and method of treating hypoxic tumors and diseases using ITPP-Ca/Na in an effective dose. Another object of the invention is to provide a composition and method 25 using ITPP-Ca/Na in an effective dose that can regulate oxygen tension in the tissue, especially a tumor. A further object of the invention is to provide a simple and easily administered, preferably oral, composition that is capable of causing significant right shifts of the P 50 value for red blood cells using ITPP-Ca/Na in an effective dose. - 12 - WO 2009/145751 PCT/US2008/006837 These and other objects, features and advantages of the present invention will become apparent after a review of the following detailed description of the disclosed embodiments. 5 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows ITPP-Ca/Na loaded red blood cell suppression of HIF-1 induction, VEGF and angiogenesis of hypoxic endothelial cells in vitro. Figure 2 shows the potential of ITPP-Ca/Na as a dual action radiation sensitzer and angiogenesis inhibitor in pancreatic and rectal cancers. 10 Figure 3 shows an agarose gel indicating the VEGF mRNA concentrations in tumors from control and ITPP drinking animals. Figure 4 show a Western blot assay of expressed VEGF in tumors of control and ITPP-treated Lewis Lung carcinoma (LLC) tumor-bearing animals. Figure 5 shows a synthesis scheme for synthesizing the monocalcium 15 tetrasodium salt of myo-inositol 1,6:2,3:4,5 tripyrophosphate. Figure 6 shows an oxygen fixation curve for hemoglobin after incubation with ITPP-Ca4Na. Figure 7 shows the percent shift of the p50 of whole human blood after incubation with ITPP-Ca4Na. 20 DETAILED DESCRIPTION OF THE INVENTION Compositions that are useful in accordance with the present invention include the mixed calcium/sodium salt of inositol-tripyrophosphate (ITPP-Ca/Na). 25 ITPP exhibits anti-angiogenic and anti-tumor properties, and is useful in controlling angiogenesis-, or proliferation-related events, conditions or substances. As used herein, the control of an angiogenic-, or proliferation-related event, condition, or substance refers to any qualitative or quantitative change in any type of factor, condition, activity, indicator, chemical or combination of chemicals, mRNA, receptor, 30 marker, mediator, protein, transcriptional activity or the like, that may be or is believed to be related to angiogenesis or proliferation, and that results from -13 - WO 2009/145751 PCT/US2008/006837 administering the composition of the present invention. Those skilled in the art will appreciate that the invention extends to other compositions or compounds in the claims below, having the described characteristics. These characteristics can be determined for each test compound using the assays detailed below and elsewhere in 5 the literature. Other such assays include counting of cells in tissue culture plates or assessment of cell number through metabolic assays or incorporation into DNA of labeled (radiochemically, for example 3 H-thymidine, or fluorescently labeled) or immuno-reactive (BrdU) nucleotides. In addition, antiangiogenic activity may be 10 evaluated through endothelial cell migration, endothelial cell tubule formation, or vessel outgrowth in ex-vivo models, such as rat aortic rings. When administered orally, ITPP exhibits anti-tumor and anti proliferative activity with little or no toxicity. ITPP was tested for its ability to induce a decrease of the 0 2 -affinity of hemoglobin measured. as a shift of the P 50 value 15 (P 5 o at 50% saturation of hemoglobin). With murine hemoglobin and whole blood, P 50 shifts to higher P0 2 of up to 250% with hemoglobin and up to 40% with whole blood were observed. The results obtained with ITPP in mice and pigs strongly suggest the possibility of its development as a therapeutic, due to its ability to enhance, in a 20 regulated manner, oxygen delivery by red blood cells in the cases of blood flow impairment. It has been found that pigs injected intravenously with ITPP-Na at a rate of lg/kg weight had beneficial properties associated with the introduction of ITPP-Na into their systems (as described in US Provisional Patent Application 25 60/585,804, which is herein incorporated by reference in its entirety); however, the introduction of ITPP-Na also resulted in a number of adverse side effects. These side effects included flushing, an increase in the heart rate, and a decrease in the Ca 2 plasma concentration. Therefore a less toxic form of ITPP that maintains a good solubility profile is needed. 30 ITPP, when administered orally, intravenously, or intraperitoneally, inhibits angiogenesis in growing tumors by enhancing P0 2 in the forming tumors. This invention further provides for methods of regulation of vascular endothelial growth factor (VEGF) in a human or animal, by administering to the human or animal - 14 - WO 2009/145751 PCT/US2008/006837 an effective amount of ITPP. More particularly, this invention provides for dose dependent effects of ITPP on VEGF mRNA and protein expressions in the LLC cell line. VEGF gene expression in tumor bearing C57BL/6 mice was assayed and the effects of ITPP-induced down regulation of VEGF have been determined and 5 correlated with modulation of cell proliferation. This invention resulted in the development of methods to control VEGF mRNA expression, protein concentration, and tumor cell proliferation. The results of these studies indicate a strong correlation between dose-dependent ITPP-induced down regulation of VEGF and cellular proliferation and suggests that ITPP can reduce VEGF mediated tumor angiogenesis, 10 as well as the rate of tumor cell proliferation. Thus, down-regulation of VEGF by ITPP decreases tumor cell proliferation. The shifting of the P 50 value to higher 0 2 -partial pressures inhibits the expression of the hypoxia gene encoding VEGF in the tumors. Expression of the hypoxia gene encoding VEGF is necessary for angiogenesis to be stimulated in 15 tumors. If this does not occur, angiogenesis is seriously inhibited and new vessels are not formed in tumors. The results obtained concerning VEGF expression suggests that oxygen partial pressure in tumors is elevated upon administration of ITPP, as this elevation is the cause of inhibition of expression of this hypoxia gene. This observation raises a 20 very important question, namely, whether this enhancement of P0 2 may act as a powerful radiosensitizer of cancer cells. Oxygen is a very potent radiosensitizer and, if indeed P0 2 in the tumors is enhanced by ITPP, this may have major consequences in enhancing the efficacy of radiation therapy of cancer. ITPP is a potential significant adjuvant in the therapy of solid tumors 25 as inhibitor of angiogenesis on one hand, and as a radiosensitizer on the other. It is known that medial temporal oxygen metabolism is markedly affected in patients with mild-to-moderate Alzheimer's disease. This measure substantiated the functional impairment of the medial temporal region in Alzheimer's disease. It also known that mean oxygen metabolism in the medial temporal, as well 30 as in the parietal and lateral temporal cortices is significantly lower in the patients that are shown to have Alzheimer's disease than in control groups without Alzheimer's disease (see Ishii et al., J. Nucl Med. 37(7):1159-65, Jul 1996, which is herein incorporated by reference in its entirety). Thus, one potential means of treating - 15 - WO 2009/145751 PCT/US2008/006837 patients shown to have Alzheimer's disease is to increase oxygen across the blood brain barrier. One method of doing so would be to use an allosteric effector of hemoglobin such as treatment with ITPP, such as with the calcium/sodium salt of ITPP. 5 The use of ITPP, such as with the calcium/sodium salt of ITPP, may also help in the treatment of a variety of vascular diseases associated with various forms of dementia. Because the brain relies on a network of vessels to bring it oxygen-bearing blood, if the oxygen supply to the brain fails, brain cells are likely to die and this can cause symptoms of vascular dementia. These symptoms can occur 10 either suddenly, following a stroke, or over time through a series of small strokes. Thus, one potential means of treating patients with vascular diseases associated with various forms of dementia is to increase the oxygen available to affected areas such as across the blood brain barrier. One method of doing so would be to use an allosteric effector of hemoglobin such as treatment with ITPP, such as with the calcium/sodium 15 salt of ITPP. Moreover, treatment of an individual with an allosteric effector of hemoglobin such as the calcium/sodium salt of ITPP may have beneficial effects for both stroke victims and osteoporosis. Although stroke and the bone-thinning disease osteoporosis are usually thought of as two distinct health problems, it has been found 20 that there may be a connection between them. Patients who survive strokes are significantly more likely to suffer from osteoporosis, a disease that puts them at high risk for bone fractures. Often, the fractures in stroke patients occur on the side of the body that has been paralyzed from the stroke. It is known that a stroke occurs when the supply of blood and oxygen to 25 the brain ceases or is greatly reduced. If a portion of the brain loses its supply of nutrient-rich blood and oxygen, the bodily functions controlled by that part of the brain (vision, speech, walking, etc.) are impaired. Annually, more than 500,000 people in the United States suffer strokes and 150,000 of those people die as a result thereof. One means of increasing oxygen flow to the brain is by use of an allosteric 30 effector of hemoglobin such as treatment with the calcium/sodium salt of ITPP. Accordingly, a potential method of treating individuals who might potentially suffer stroke or osteoporosis is by treatment of an individual with, for example, the calcium/sodium salt of ITPP. - 16 - WO 2009/145751 PCT/US2008/006837 Also contemplated by the present invention are implants or other devices comprised of the compounds or drugs of ITPP, or prodrugs thereof, where the drug or prodrug is formulated in a biodegradable or non-biodegradable polymer for sustained release. Non-biodegradable polymers release the drug in a controlled 5 fashion through physical or mechanical processes without the polymer itself being degraded. Biodegradable polymers are designed to gradually be hydrolyzed or solubilized by natural processes in the body, allowing gradual release of the admixed drug or prodrug. The drug or prodrug can be chemically linked to the polymer or can be incorporated into the polymer by admixture. Both biodegradable and non 10 biodegradable polymers and the process by which drugs are incorporated into the polymers for controlled release are well known to those skilled in the art. Examples of such polymers can be found in many references, such as Brem et al., J. Neurosurg 74: pp. 441-446 (1991), which is herein incorporated by reference in its entirety. These .implants or devices can be implanted in the vicinity where delivery is desired, 15 for example, at the site of a tumor. In addition to the compounds of the present invention, the pharmaceutical composition of this invention may also contain, or be co-administered (simultaneously or sequentially) with, one or more pharmacological agents of value in treating one or more disease conditions referred to hereinabove. 20 A person skilled in the art will be able by reference to standard texts, such as Remington's Pharmaceutical Sciences 17 'h edition, to determine how the formulations are to be made and how these may be administered. In a further aspect of the present invention there is provided use of compounds of ITPP, such as ITPP-Ca/Na or prodrugs thereof, according to the present 25 invention for the preparation of a medicament for the prophylaxis or treatment of conditions associated with angiogenesis or accelerated cell division or inflammation. In a further aspect of the present invention there is provided a pharmaceutical composition comprising compounds of ITPP, such as ITPP-Ca/Na or prodrugs thereof, according to the present invention, together with a pharmaceutically 30 acceptable carrier, diluent, adjuvant or excipient. The pharmaceutical composition may be used for the prophylaxis or treatment of conditions associated with angiogenesis or accelerated cell division or -17- WO 2009/145751 PCT/US2008/006837 inflammation, for treatment of Alzheimer's disease, treatment of stroke and/or osteoporosis. In a still further aspect of the present invention there is provided a method of prophylaxis or treatment of a condition associated with angiogenesis or 5 accelerated or increased amounts of cell division, hypertrophic growth, or inflammation, said method including administering to a patient in need of such prophylaxis or treatment an effective amount of compounds of ITPP, such as ITPP Ca/Na or prodrugs thereof, according to the present invention, as described herein. It should be understood that prophylaxis or treatment of said condition includes 10 amelioration of said condition. By "an effective amount" as referred to in this specification, it is meant a therapeutically or prophylactically effective amount. Such amounts can be readily determined by an appropriately skilled person, taking into account the condition to be treated, the route of administration and other relevant factors. Such a person will 15 readily be able to determine a suitable dose, mode and frequency of administration. "Individual" as referred to in this application refers to any animal that may be in need of treatment for a given condition. "Individual" includes humans, other primates, household pets, livestock, rodents, other mammals, and any other animal(s) that may typically be treated by a veterinarian. 20 The compositions described above can be provided as physiologically acceptable formulations using known techniques, and these formulations can be administered by standard routes. In general, the combinations may be administered by the topical, oral, rectal, intraperitoneal or parenteral (e.g., intravenous, subcutaneous or intramuscular) route. In addition, the combinations may be incorporated into 25 polymers allowing for sustained release, the polymers being implanted in the vicinity of where delivery is desired, for example, at the site of a tumor, or into an a cavity or blood vessel that will lead to easy delivery to the place to be treated. The dosage of the composition will depend on the condition being treated, the particular derivative used, and other clinical factors such as weight and condition of the patient and the 30 route of administration of the compound. However, for oral administration, a recommended dosage is in the range of 0.1 to 5.0 g/kg/day. A dosage for oral administration is in the range of 0.5 to 2.0 g/kg/day or alternatively, about 0.5 to about 1.5 g/kg/day. In an alternate embodiment, a dosage for oral administration is in -18- WO 2009/145751 PCT/US2008/006837 the range of about 0.80 to 1.0 g/kg/day or alternatively, about between 0.9 to 1.1 g/kg/day. The formulations in accordance with the present invention can be administered in the form of tablet, a capsule, a lozenge, a cachet, a solution, a 5 suspension, an emulsion, a powder, an aerosol, a suppository, a spray, a pastille, an ointment, a cream, a paste, a foam, a gel, a tampon, a pessary, a granule, a bolus, a mouthwash, or a transdermal patch. The formulations include those suitable for oral, rectal, nasal, inhalation, topical (including dermal, transdermal, buccal and sublingual), vaginal, 10 parenteral (including subcutaneous, intramuscular, intravenous, intraperitoneal, intradermal, intraocular, intratracheal, and epidural) or inhalation administration. The formulations may conveniently be presented in unit dosage form and may be prepared by conventional pharmaceutical techniques. Such techniques include the step of bringing into association the active ingredient and a pharmaceutical carrier(s) or 15 excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. Formulations contemplated as part of the present invention include nanoparticles formulations made by methods disclosed in U.S. Patent Application No. 20 10/392,403 (Publication No. 2004/0033267) which is hereby incorporated by reference in its entirety. By forming nanoparticles, the compositions disclosed herein are shown to have increased bioavailability. Preferably, the particles of the compounds of the present invention have an effective average particle size of less than about 2 microns, less than about 1900 nm, less than about 1800 nm, less than about 1700 nm, less than about 1600 nm, less than about 25 1500 nm, less than about 1400 nm, less than about 1300 nm, less than about 1200 nm, less than about 1100 nm, less than about 1000 nm, less than about 900 nm, less than about 800 run, less than about 700 nm, less than about 600 nm, less than about 500 nm, less than about 400 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 100 nm, less than about 75 nm, or less than about 50 nm, as measured by 30 light-scattering methods, microscopy, or other appropriate methods well known to those of ordinary skill in the art. Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing - 19 - WO 2009/145751 PCT/US2008/006837 a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in water liquid emulsion or a water-in-oil emulsion, etc. A tablet may be made by compression or molding, optionally with one 5 or more accessory ingredients. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Molded tablets may be made by molding, in a suitable machine, a mixture of the powdered compound moistened with an inert liquid 10 diluent. The tablets may optionally be coated or scored and may be formulated so as to provide a slow or controlled release of the active ingredient therein. Formulations suitable for topical administration in the mouth include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin 15 and glycerin, or sucrose and acacia; and mouthwashes comprising the ingredient to be administered in a suitable liquid carrier. Formulations suitable for topical administration to the skin may be presented as ointments, creams, gels and pastes comprising the ingredient to be administered in a pharmaceutically acceptable carrier. A preferred topical delivery 20 system is a transdermal patch containing the ingredient to be administered. Formulations for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter and/or a salicylate. Formulations suitable for nasal administration, wherein the carrier is a 25 solid, include a coarse powder having a particle size, for example, in the range of 20 to 500 microns which is administered in the manner in which snuff is taken; i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable formulations, wherein the carrier is a liquid, for administration, as for example, a nasal spray or as nasal drops, include aqueous or 30 oily solutions of the active ingredient. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing, in -20- WO 2009/145751 PCT/US2008/006837 addition to the active ingredient, ingredients such as carriers as are known in the art to be appropriate. Formulation suitable for inhalation may be presented as mists, dusts, powders or spray formulations containing, in addition to the active ingredient, 5 ingredients such as carriers as are known in the art to be appropriate. Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may 10 include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in freeze-dried lyophilizedd) conditions requiring only the addition of a sterile liquid carrier, for example, water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile 15 powders, granules and tablets of the kinds previously described. Preferred unit dosage formulations are those containing a daily dose or unit, daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the administered ingredient. It should be understood that in addition to the ingredients, particularly 20 those mentioned above, the formulations of the present invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents or other agents to make the formulation more palatable and more easily swallowed. 25 Experimental For the in vitro experiments, ITPP was dissolved in deionized water, pH was adjusted at pH 7 and, for incubation with whole blood, the osmolarity of the ITPP solutions was adjusted with glucose to 270-297 mOsM. Mixtures of hemoglobin and ITPP were measured with a HEMOX analyzer (PD Marketing, London) 30 immediately after mixing. Red blood cells were incubated with ITPP for 1 hour at 37*C. Following incubation, the cells were washed 3 times with Bis-Tris-buffer (pH=7.0) and then used for Pso measurement. -21 - WO 2009/145751 PCT/US2008/006837 In experiments conducted in vivo in which ITPP was administered orally, a significant shift of the P 5 0 value of circulating RBCs was observed. ITPP was dissolved in drinking water at a 20g/L-concentration (.27mM, pH -7.0.) and offered for drinking ad libitum. 5 The following examples illustrate but do not limit the invention. Thus, the examples are presented with the understanding that modifications may be made and still be within the spirit and scope of the invention. EXAMPLE 1 10 Effectiveness of the Mixed Calcium Sodium Salt of myo-Inositol Tripyrophosphate When myo-inositol tripyrophosphate-sodium salt (ITPP-Na) is mixed with CaCI 2 , a mixture of ITPP-Na (myo-inositol tripyrophosphate-sodium salt) and ITPP-Ca (myo-inositol tripyrophosphate-calcium salt) is obtained. This mixture, when added to free hemoglobin or to whole blood induces a PSo shift of 170% and 15 25%, respectively as shown in Tables 2 and 3 below. Please see the results in Tables 2 and 3 for compound 15. The compounds in Tables 2 and 3 are as follows: 4 is the pyridinium salt of ITPP, 5 is the sodium salt of ITPP (i.e., ITPP-Na), 7 is the N,N dimethylcyclohexyl ammonium salt of ITPP, 11 is the cycloheptyl ammonium salt of ITPP, 12 is the cyclooctyl ammonium salt of ITPP, 13 is the piperazinium salt of 20 ITPP, 14 is the tripiperazinium salt of ITPP, and 15 is the calcium salt of ITPP (i.e., ITPP-Ca). In Tables 2 and 3, the effectiveness of all of the salts of ITPP regarding their ability to act as allosteric effectors of hemoglobin can be seen. The sodium salt and the calcium salt of ITPP appear to be the best allosteric effectors for both free 25 hemoglobin (Table 2) and in whole blood (Table 3). However, pigs injected intravenously with ITPP-Na at a rate of lg/kg weight resulted in a number of adverse side effects. The intravenous injection of pigs with ITPP-Na resulted in flushing, an increase in the heart rate, and a decrease in the Ca2+ plasma concentration from 2.38 mmol/L to 1.76 mmol/L. 30 Administration of the mixture of the sodium and calcium salt of ITPP, at the same dosage did not induce any of the cited effects and the Ca 2 + plasma concentration stayed unchanged at 2.38 mmol/L. -22 - WO 2009/145751 PCT/US2008/006837 Figure 1 shows the ability of ITPP-Ca/Na loaded red blood cells suppression of HIF-1 induction, VEGF and angiogenesis of hypoxic endothelial cells in vitro. Figure 2 demonstrates the potential of ITPP-Ca/Na as a radiation sensitizer and as an angiogenesis inhibitor in pancreatic and rectal cancers. 5 Table 2. P 50 values of free Hb after incubation with compounds 4, 5, 7, 11-14 and 15, in vitro
P
5 o (Torr) P 50 (Torr) P 50 increase Compound Free Hb Hb + compound (%) + SD 4 (H) 15.3 31.6 107 ± 22 (M) 25.0 50.0 100 ±18 5 (H) 15.3 49.8 225 ±19 (M) 24.9 69.7 180 ±25 (P) 22.0 68.1 209 ±39 7 (M) 24.9 45.1 81 ±15 11 (M) 24.9 43.8 71± 3 12 (M) 24.9 30.6 23± 5 13 (M) 23.4 67.7 189 ±43 14 (M) 23.4 82.9 254 ± 49 15 (H) 123 33.1 170 ± 32 (M) 26.9 61.9 130 ± 30 H = human; M = murine; P = porcine free Hb. Concentration of the compound solution was 60 mM; means of P 50 shifts in % are shown. SD = standard deviation. Compounds 4, 7, 11, 12, 14 and 15: 10 three P 5 0 values each were used for the calculation of means; compound 5: with human blood: five values, murine blood: ten values and porcine blood; three values were used for the calculation of the means of P 50 shifts in %. -23 - WO 2009/145751 PCT/US2008/006837 Table 3. P 50 values of whole blood after incubation with compounds 4, 5, 7, 11-14 and 15, in vitro
P
50 (Torr)
P
50 (Torr) compound + P 50 increase Compound whole blood whole blood (%) + SD 4 (H) 22.1 24.3 10± 4 (M) 37.9 42.7 13± 2 5 (H) 22.1 30.8 39a ±5 (P) 31.6 44.2 40a ±3 (M) 36.7 47.4 29 b 3 7 (M) 40.1 52.0 30± 3 11 (M) 37.9 45.5 20± 2 12 (M) 37.9 41.3 9± 1 13 (M) 37.9 41.7 10± 2 14 (M) 39.2 41.9 7± 1 15 (M) 39.2 42.3 8 ± 2 (H) 24.8 31.0 25± 3 (M) 40.1 55.3 38a ±4 H = human; M = murine; P = porcine whole blood. Compound concentrations: 30 mM; means of (four single values) Pso shifts E SD are shown. a Compound concentration: 60 mM. b Compound concentration: 4 mM. 5 EXAMPLE 2 10 Effect of in vivo Lowering of Hemoglobin's Affinity for 02 by ITPP on Intratumoral P02 Angiogenesis and Expression of VEGF mRNA ITPP, when administered orally, intravenously, or intraperitoneally, inhibits angiogenesis in growing tumors by enhancing the P0 2 in the forming tumors. Thirty (30) C57BL/6 mice received 20 g/L of ITPP orally until the P 5 0 value showed a 15 shift of at least 20% above the control value. Thereafter, all animals received Ix10 6 Lewis Lung carcinoma (LLC) cells, injected in the dorsal cavity. At different time points, the VEGF mRNA were assayed by RT-PCR in the tumors growing in both groups of mice. - 24 - WO 2009/145751 PCT/US2008/006837 Tumor tissue samples were ground in a RIPA lysis buffer (1% Nonidet p-40 detergent, 50mM Tris pH 8.0, 137mM NaCI, 10% glycerol) supplemented with protease inhibitor cocktail (Roche, Reinach, Switzerland). After centrifugation for 10 minutes at 4*C and 12,000 g, protein concentrations of tissue extracts were 5 determined according to the Bradford method. Detergent soluble protein samples (10 mg) were separated by size on a SDS-PAGE in 10% acrylamide gels and transferred to nitrocellulose membrane (Protran BA 85, Schleicher and Schuell, Dassel, Germany). Membranes were blocked for 3 hours at room temperature in 10% skim milk in Tris buffer saline containing 0.1% Tween, before an overnight incubation at 4*C with 10 rabbit polyclonal antibodies recognizing human, mouse and rat vascular endothelial growth factor (VEGF A-20, sc-152, Santa Cruz Biotechnology, Santa Cruz, California) at a dilution of 1:200. Membranes were then probed for primary antibody with anti-rabbit (1:16,000) peroxidase conjugates (Sigma-Aldrich, L'Isle d'Abeau Chesnes, France) for 60 minutes at room temperature. The resulting complexes were 15 visualized by enhanced chemiluminescence autoradiography (Amersham Pharma Biotech, Orsay, France). There was a difference in the level of mRNA of the VEGF gene in both groups. Figure 3 shows an agarose gel indicating the VEGF mRNA concentrations in tumors from control and ITPP drinking animals. The RT-PCR agarose gel assay of 20 VEGF mRNAs from tumor tissue taken from 2 mice each on day 15 after inoculation of LLC cells (track 1: controls, track 2: ITPP treated animals) and day 30 after inoculation (track 3: control animals, track 4: ITPP treated animals). Figure 4 shows the Western blot assay of the expressed VEGF in tumors of control and ITPP-treated LLC tumor-bearing animals. 25 Quantification of the gel assays indicated a reduction by a factor of 10,000 of the amount of VEGF mRNAs detected in the tumors of animals having received ITPP, at day 9 and then, while differences remain between treated and untreated animals, they tend to decrease. This indicates that ITPP taken up by circulating red blood cells significantly increases tumor P0 2 . 30 EXAMPLE 3 Method of Synthesizing Monocalcium Tetrasodium myo-Inositol Tripyrophosphate -25 - WO 2009/145751 PCT/US2008/006837 Materials: 1. myo-Inositol hexakisphosphate dodecasodium salt (Product Number: P0109, Sigma). 2. Dicyclohexylcarbodiimide (Product Number: D80002, Aldrich). 5 3. Triethylamine (Product Number: 15791, Acros) 4. Dowex 50Wx8 hydrogen form (Product Number: 217506, Aldrich). 5. Ca(OH) 2 (Product Number: 239232, Aldrich). 6. NaOH (Product Number: 1040017, Sds). 7. Acetonitrile (Product Number: 34851, Aldrich). 10 8. Deionized Water. Procedure: The following synthesis scheme is shown in Figure 5. Dowex 50WX8 200 ion exchange resin (800 g) was washed with water until the elute was colorless. myo-Inositol hexakisphosphate dodecasodium salt (note-1) (100 g, 0.108 mol, 1.0 eq) 15 was added portionwise (10 g/portion in about 45 minutes) to 500 mL of water. Each portion was dissolved with stirring at room temperature (23 *C) before the next portion was added. This solution was then passed through the column containing the above washed Dowex 50WX8-200 ion exchange resin and eluted with water (4x200 mL) to obtain the free phytic acid (note-2). To the combined acidic fractions, 20 triethylamine (400 mL, 2.87 mol, 26.5 eq, about twice the theoretical quantity) was added over 1 to 2 minutes at room temperature (23 *C) and the mixture was stirred vigorously for 15 minutes (note-3). Then the solvent was evaporated on a rotary evaporator (60 *C, 68-22 mbar) (note-4) and the residue was dried under high vacuum for 1 hr at room temperature (23 *C) to give the hexatriethylammonium myo-inositol 25 hexakisphosphate (note-5). To this hexatriethylammonium myo-inositol hexakisphosphate dissolved in water (800 mL), dicyclohexylcarbodiimide (142 g, 0.68 mol, 6.3 eq) dissolved in acetonitrile (1600 mL) was added at once and the mixture was refluxed for 12 h (note 6). One more equivalent of dicyclohexylcarbodiimide (22 g, 0.108 mol, 1.0 eq) 30 dissolved in acetonitrile (40 mL) was added and refluxed for further 6 h (note-7). The mixture was cooled to room temperature (23 *C) and the dicyclohexylurea formed was -26 - WO 2009/145751 PCT/US2008/006837 filtered through a sintered funnel (note-8) and washed with water (3x200 mL). The filtrate was evaporated on a rotary evaporator (60 *C, 68-22 mbar) and dried under high vacuum at room temperature (23 *C) (note-9). The resulting sticky syrupy residue was redissolved in 400 mL of water to remove all dicyclohexylurea that had 5 remained dissolved in acetonitrile, filtered through a sintered funnel (note-8), and washed with water (2x100 mL). The filtrate was evaporated on a rotary evaporator (60 *C, 68-22 mbar) and dried under high vacuum at room temperature (23 *C). The resulting residue was redissolved in 200 mL of water to remove any further dicyclohexylurea that had remained dissolved in solution, filtered through a sintered 10 funnel (note-8), and washed with water (2x100 mL). The filtrate was evaporated on a rotary evaporator (60 *C, 68-22 mbar) and dried under high vacuum at room temperature (23 'C) affording hexatriethylammonium myo-inositol 1,6:2,3:4,5 trispyrophosphate (note- 10). This hexatriethylammonium myo-inositol 1,6:2,3:4,5 trispyrophosphate 15 salt was dissolved in 400 mL of water, passed through a column (note-I 1) containing prewashed Dowex 50WX8-200 (400 g) ion exchange resin and eluted with water (4x100 mL) (note-12). To the combined acidic fractions was immediately added solid Ca(OH) 2 (5.56 g, 0.075 mol, 1.0 eq) followed by addition of a NaOH solution [(12.0 g, 0.300 mol, 4.0 eq) in 25 mL of water)] at room temperature (23 *C).(note-13). Then 20 the pH of the reaction mixture was carefully adjusted to around 6.99 with a solution of 1:4 Ca(OH) 2 :NaOH (1.5 g of Ca(OH) 2 and 3.23 g of NaOH in 1500 mL of water, ~ 1385 mL brought the pH to ~ 6.99) (note-14). Finally, the solvent was evaporated on a rotary evaporator (60 *C, 68-22 mbar) and dried under high vacuum at room temperature (23 *C) to yield the monocalcium tetrasodium myo-inositol 1,6:2,3:4,5 25 trispyrophosphate, ITPP Ca4Na (77.2 g, 97%) as a white solid. The compound obtained has been characterized by proton and phosphorous-31 NMR spectroscopy, mass spectroscopy, elemental analysis, cation determination by atomic absorption and water content. It contains less than 2% other phosphorous compounds. Elemental analysis (ICP atomic absorption): P 20%; Ca 30 4.2%; Na 10.3% (calc. : P 25.4%; Ca 5.5%; Na 12.6%). Water content: about 18-23% depending on drying conditions. Notes: -27 - WO 2009/145751 PCT/US2008/006837 Note-1. Purity checked in-house by 'H and "P NMR (>98%), as well as HPLC, elemental analysis and atomic absorption for cation determination. Note-2. Collect the elutes which are acidic (pH paper). When all phytic acid is eluted, the elute becomes neutral. 5 Note-3. Addition of triethylamine generates some heat. Add progressively. Note-4. Lower temperature can be used if evaporation can be achieved with the equipment available. Note-5. Purity and characterization was checked by 'H and "P NMR. Note-6. Reflux temperature was about 80 *C. Heated with a mantle. 10 Note-7. After 12 h, more than 98% product has been formed. Addition of more dicyclohexylcarbodiimide led to >99% product formation. Note-8. The porosity of the sintered funnel used was 4 and this will effectively filter off the dicyclohexylurea byproduct. Note-9. Thorough drying is necessary in order to be able to remove all the remaining 15 dicyclohexylurea byproduct. Note-10. Characterized by 'H and 31 P NMR, purity >99%. Note- 11. Two typical procedures are for instance: Procedure 1: The column frit porosity was 1. The diameter of the column was 8 cm and the 20 length of the Dowex bed was 9.5 cm. The solution was eluted first in 15 minutes without any pressure and then the washings under some pressure were eluted within 5 minutes. Procedure 2: The column frit porosity was 2. The diameter of the column was 6 cm and the 25 length of the Dowex bed was 16.5 cm. The solution was eluted first in 45 minutes without any pressure and then the washings under some pressure were eluted within 5 10 minutes. On the basis of numerous preparations, the following is recommended: a) a column frit porosity of 1. -28- WO 2009/145751 PCT/US2008/006837 b) a Dowex column with a height/diameter ratio of 1.5-2.0, so that the elution time be less than 30 minutes. If the elution is too slow then flush with some pressure. Note-12. CAUTION: As the myo-inositol 1, 6:2,3:4,5 trispyrophosphate free acid may 5 hydrolyse after standing for a long time at low pH (< 1.0), the pH should be adjusted quickly to about 3-4 in order to avoid any such hydrolysis. Check then for absence of triethylamine signals in 'H NMR (4-1 ppm) and for absence of phosphorous signals around 2-4 ppm. In the unlikely case that the 'H NMR shows the presence of triethylamine, the whole solution has to be 10 passed again over a fresh Dowex-H* in order to remove it. It is very important that there be no triethylamine left, as it would remain in the final material. Note- 13. It is very important to add first solid Ca(OH) 2 and make sure that it is 15 completely dissolved. After addition of the NaOH solution, the pH of the reaction mixture was ~ 1.6. The total amount of Ca(OH) 2 :NaOH required to neutralize the reaction mixture to pH ~ 6.9 was - 6.9:14.9 g, respectively. In order to minimize the amount of 1:4 Ca(OH) 2 :NaOH solution required at the end (and reduce the final volume), add initially 6.3 g of solid Ca(OH) 2 20 followed by 13.6 g of NaOH ( in 25 mL of water). Thereafter, the amount of 1:4 Ca(OH) 2 :NaOH solution required to bring the pH to. 6.9-7.0 will be significantly reduced. Note-14. The 1:4 Ca(OH) 2 :NaOH solution should be freshly prepared and well closed; 25 otherwise, CO 2 from the atmosphere will be absorbed and insoluble materials will be formed. Adjust the pH of the solution close to 7 and do not go beyond 7. - 29 - WO 2009/145751 PCT/US2008/006837 EXAMPLE 4 Biological Activity of the Monocalcium Tetrasodium Salt of myo-Inositol Tripyrophosphate (ITPP-Ca4Na) ITPP-Ca4Na acts as a powerful effector of hemoglobin shifting the oxygen 5 fixation curves to the right, with respect to the natural effector bisphosphoglycerate. The p50 values increase with concentration as show in Figures 6 and 7. In Figure 6, hemoglobin is incubated (at concentrations up to 100 mM final) with ITPP Ca4Na, for 1 hour at 37"C and measured by TCS-hemox analyzer for p50 shifts. In Figure 7, whole human blood was incubated (at concentrations up to 120 mM final) with ITPP Ca4Na, 10 for 1 hour 37'C and measured by TCS-hemox analyzer for p50 shifts. Having described the invention with reference to particular compositions, method for detection, and source of activity, and proposals of effectiveness, and the like, it will be apparent to those of skill in the art that it is not 15 intended that the invention be limited by such illustrative embodiments or mechanisms, and that modifications can be made without departing from the scope or spirit of the invention, as defined by the appended claims. It is intended that all such obvious modifications and variations be included within the scope of the present invention as defined in the appended claims. It should be understood that any of the 20 above described one or more elements from any embodiment can be combined with any one or more element in any other embodiment. Moreover, when a range is mentioned, it should be understood that it is contemplated that any real number that falls within the range is a contemplated end point. For example, if a range of 0.9 and 1.1 g/kg is given, it is contemplated that any real number value that falls within that 25 range (for example, 0.954 to 1.052 g/kg) is contemplated as a subgenus range of the invention, even if those values are not explicitly mentioned. All references referred to herein are incorporated by reference in their entireties. Finally, the above description is not to be construed to limit the invention but the invention should rather be defined by the below claims. - 30 - WO 2009/145751 PCT/US2008/006837 REFRENCES 1. Fylaktakidou, K., Lehn, J.-M., Greferath, R., and Nicolau, C. (2004) Bioorg. Med. Chem. Lett (submitted) 2. Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips BE, Ferrara N (1993) Nature 362, 841-844. 3. Kandel J, Bossy-Wetzel E, Radvanyi F, Klagsbrun M, Folkman J, Hanahan D (1991) Cell 66, 1095-1104. 4. O'Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn B, Birkhead JR. Olsen BR, Folkman J (1997) Cell 88, 277-285. 5. Good DJ, Polverini PJ, Rastinejad F, Le Beau mm, Lemons RS, Frazier WA, Bouck NP. (1990) Proc Natl Acad Sci USA 87, 6624-6628. 6. O'Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal R.A, Moses M, Lane WS, Cao Y, Sage EH, Folkman J (1994) Cell 79, 3 15-328. 7. Chen C, Parangi S, Tolentino MJ, Folkman J. (1995) Cancer Res. 55, 4230-4233. 8. Ferrara N. (2002) Nat. Rev. Cancer 2, 795-803. 9. Ferrara N, Davis-Smyth T (1997) Endocr Rev. 18, 4-25. 10. Ferrara N, Gerber HP, LeCouter J. (2003) Nat Med. 9, 669-676. 11. Fontanini G, Vignati S, Boldrini L, Chine S, Silvestri V, Lucchi M, Mussi A, Angeletti CA, Bevilacqua G. (1997) Clin Cancer Res. 3, 861-865. 12. Dory, Porat R, Keshet E. (2001) Am J Physiol Cell Physiol. 280, C1367-1374. 13. Brizel DM, Scully SP, Harrelson JM, Layfield U, Bean JM, Prosnitz LR, Dewhirst MW (1996) Cancer Res. 56, 94 1-943. 31

Claims (30)

1. A monocalcium tetrasodium salt of inositol tripyrophosphate.
2. The monocalcium tetrasodium salt of inositol tripyrophosphate of claim 1, wherein the inositol tripyrophosphate is myo-inositol 1,6:2,3:4,5 tripyrophosphate.
3. A pharmaceutical composition comprising a monocalcium tetrasodium salt of inositol tripyrophosphate and a pharmaceutically acceptable adjuvant, diluent, carrier, or excipient thereof.
4. The pharmaceutical composition of claim 3, wherein the inositol tripyrophosphate is myo-inositol 1,6:2,3:4,5 tripyrophosphate.
5. The pharmaceutical composition of claim 4, wherein the myo-inositol 1,6:2,3:4,5 tripyrophosphate is prepared at an effective dosage for treating cancer in a subject.
6. The pharmaceutical composition of claim 5, wherein the dosage of myo-inositol 1,6:2,3:4,5 tripyrophosphate is between 0.5 and 1.5 g/kg.
7. The pharmaceutical composition of claim 5, wherein the dosage of myo-inositol 1,6:2,3:4,5 tripyrophosphate is between 0.9 and 1.1 g/kg.
8. The pharmaceutical composition of claim 5, wherein the cancer is selected from the group consisting of rhabdomyosarcomas, retinoblastoma, Ewing's sarcoma, neuroblastoma, and osteosarcoma. - 32 - WO 2009/145751 PCT/US2008/006837
9. The pharmaceutical composition of claim 5, wherein the cancer is selected from one or more of the group consisting of breast cancer, prostate cancer, renal cell cancer, brain cancer, ovarian cancer, colon cancer, bladder cancer, pancreatic cancer, stomach cancer, esophageal cancer, cutaneous melanoma, liver cancer, lung cancer, testicular cancer, kidney cancer, bladder cancer, cervical cancer, lymphoma, parathyroid cancer, penile cancer, rectal cancer, small intestine cancer, thyroid cancer, uterine cancer, Hodgkin's lymphoma, lip and oral cancer, skin cancer, leukemia, and multiple myeloma.
10. A method of making a monocalcium tetrasodium salt of myo-inositol 1,6:2,3:4,5 tripyrophosphate comprising; a) passing a myo-inositol hexakisphosphate salt over an ion exchange column to obtain a free inositol tripyrophosphate acid, b) reacting the free myo-inositol tripyrophosphate acid with triethylamine to form a hexatriethylammonium myo-inositol hexakisphosphate, c) reacting the hexatriethylammonium myo-inositol hexakisphosphate with dicyclohexylcarbodiimide dissolved in acetonitrile to form a hexatriethylammonium myo-inositol 1,6:2,3:4,5 trispyrophosphate, d) passing the hexatriethylammonium myo-inositol 1,6:2,3:4,5 trispyrophosphate over an ion-exchange column followed by addition of a calcium salt containing organic compound and a sodium salt containing organic compound to form the monocalcium tetrasodium salt of myo-insotiol 1,6:2,3:4,5 tripyrophosphate.
11. The method of claim 10, wherein the calcium salt containing organic compound is calcium hydroxide and the sodium salt containing organic compound is sodium hydroxide.
12. A method of treating cancer comprising administering to an individual a pharmaceutically effective amount of the composition of claim 3.
13. The method of claim 12, wherein the composition is administered to an individual at a dosage of about 0.5 to about 1.5 g/kg. - 33 - WO 2009/145751 PCT/US2008/006837
14. The method of claim 12, wherein the composition is administered to an individual at a dosage of about 0.9 to about 1.1 g/kg.
15. A method of shifting the hemoglobin P 50 level towards higher values of oxygen partial pressure comprising administering to an individual an effective amount of the composition of claim 3.
16. The method of claim 15, wherein the inositol tripyrophosphate is myo-inositol 1,6:2,3:4,5 tripyrophosphate.
17. The method of claim 15, wherein the composition is administered at a dosage of about 0.5 to about 1.5 g/kg.
18. The method of claim 15, wherein the composition is administered at a dosage of about 0.9 to about 1.1 g/kg.
19. A method of treating Alzheimer's disease comprising administering to an individual an effective amount of the composition of claim 3.
20. The method of claim 19, wherein the inositol tripyrophosphate is myo-inositol 1,6:2,3:4,5 tripyrophosphate.
21. The method of claim 19, wherein the composition is administered at a dosage of about 0.5 to about 1.5 g/kg.
22. The method of claim 19, wherein the composition is administered at a dosage of about 0.9 to about 1.1 g/kg. - 34 - WO 2009/145751 PCT/US2008/006837
23. A method of enhancing the partial pressure of oxygen in hypoxic tumors comprising administering to an individual an effective amount of the composition of claim 3.
24. The method of claim 23, wherein the inositol tripyrophosphate is myo-inositol 1,6:2,3:4,5 tripyrophosphate.
25. The method of claim 23, wherein the composition is administered at a dosage of about 0.5 to about 1.5 g/kg.
26. The method of claim 23, wherein the composition is administered at a dosage of about 0.9 to about 1.1 g/kg.
27. A method of inhibiting angiogenesis in hypoxic tumors comprising administering to an individual an effective amount of the composition of claim 3.
28. The method of claim 27, wherein the inositol tripyrophosphate is myo-inositol 1,6:2,3:4,5 tripyrophosphate.
29. The method of claim 27, wherein the composition is administered at a dosage of about 0.5 to about 1.5 g/kg.
30. The method of claim 27, wherein the composition is administered at a dosage of about 0.9 to about 1.1 g/kg. -35 -
AU2008356891A 2008-05-30 2008-05-30 Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin Abandoned AU2008356891A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2008/006837 WO2009145751A1 (en) 2008-05-30 2008-05-30 Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin

Publications (1)

Publication Number Publication Date
AU2008356891A1 true AU2008356891A1 (en) 2009-12-03

Family

ID=41377364

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2008356891A Abandoned AU2008356891A1 (en) 2008-05-30 2008-05-30 Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin

Country Status (3)

Country Link
AU (1) AU2008356891A1 (en)
CA (1) CA2725788A1 (en)
WO (1) WO2009145751A1 (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003231157C1 (en) * 2002-04-29 2009-02-26 Normoxys, Inc. Inositol pyrophosphates, and methods of use thereof
US20060241086A1 (en) * 2005-03-18 2006-10-26 Claude Nicolau Calcium salt of myo-inositol 1,6:2,3:4,5 tripyrophosphate as an allosteric effector of hemoglobin

Also Published As

Publication number Publication date
WO2009145751A1 (en) 2009-12-03
CA2725788A1 (en) 2009-12-03

Similar Documents

Publication Publication Date Title
EP1863495B1 (en) Calcium and sodium salt of myo-inositol 1,6:2,3:4,5 tripyrophosphate for treating cancer
US20120010174A1 (en) Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin
US20120035137A1 (en) Use of inositol-tripyrophosphate in treating tumors and diseases
US20170027966A1 (en) Tumor eradication by inositol-tripyrophosphate
EP0110498B1 (en) Artificial lung surfactant and remedy for respiratory distress syndrome containing it as active principle
US20060106000A1 (en) Use of inositol-tripyrophosphate in treating tumors and diseases
FI79946C (en) FOERFARANDE FOER FRAMSTAELLNING AV FOSFOLIPID-SAMMANSAETTNING ANVAENDBARA VID BEHANDLING AV SJUKDOMAR I DET CENTRALA NERVSYSTEMET.
US20100029594A1 (en) Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin
JPH02191220A (en) Drug containing inositol triphosphate against abnormal level of lipoprotein
US20140194391A1 (en) Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin
EP1973398B1 (en) Use of inositol-tripyrophosphate in treating tumors mediated by angiogenesis
AU2007340371A1 (en) Cyclitols and their derivatives and their therapeutic applications
JP2005515969A (en) Vitamin storage regulation
US20120003161A1 (en) Use of inositol-tripyrophosphate in treating tumors and diseases
AU2008356891A1 (en) Calcium/sodium salt of inositol tripyrophosphate as an allosteric effector of hemoglobin
AU2002333019B2 (en) Modulation of vitamin storage
ZA200401126B (en) Modulation of vitamin storage.
AU2002333019A1 (en) Modulation of vitamin storage

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period