WO1994026761A1 - Traitement de la toxicite des agents chimioterapeutiques et des agents antiviraux avec des nucleosides de pyrimidine acyles - Google Patents

Traitement de la toxicite des agents chimioterapeutiques et des agents antiviraux avec des nucleosides de pyrimidine acyles Download PDF

Info

Publication number
WO1994026761A1
WO1994026761A1 PCT/US1993/012689 US9312689W WO9426761A1 WO 1994026761 A1 WO1994026761 A1 WO 1994026761A1 US 9312689 W US9312689 W US 9312689W WO 9426761 A1 WO9426761 A1 WO 9426761A1
Authority
WO
WIPO (PCT)
Prior art keywords
uridine
pyrimidine nucleoside
acyl derivative
cytidine
derivative
Prior art date
Application number
PCT/US1993/012689
Other languages
English (en)
Inventor
Reid Warren Von Borstel
Michael Kevin Bamat
Original Assignee
Pro-Neuron, 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 Pro-Neuron, Inc. filed Critical Pro-Neuron, Inc.
Priority to AU60812/94A priority Critical patent/AU6081294A/en
Publication of WO1994026761A1 publication Critical patent/WO1994026761A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids

Definitions

  • This invention relates generally to treatment of chemotherapeutic agent and antiviral agent toxicity with acylated derivatives of non-methylated pyrimidine
  • nucleosides are capable of attenuating damage to the hematopoietic system in animals receiving antiviral or antineoplastic chemotherapy.
  • This invention also relates to protection of other tissues affected by antiviral or antineoplastic chemotherapy, including the gastrointestinal epithelium.
  • a major complication of cancer chemotherapy and of antiviral chemotherapy is damage to bone marrow cells or suppression of their function. Specifically, chemotherapy damages or destroys hematopoietic precursor cells, primarily found in the bone marrow and spleen, impairing the production of new blood cells (granulocytes, lymphocytes, erythrocytes, monocytes, platelets, etc.). Treatment of cancer patients with 5-fluorouracil, for example, reduces the number of leukocytes
  • lymphocytes and/or granulocytes can result in enhanced susceptibility of the patients to infection. Many cancer patients die of infection or other
  • Chemotherapeutic agents can also result in subnormal formation of platelets which produces a
  • antineoplastic or antiviral chemotherapy agents act by inhibiting nucleotide biosynthesis, metabolism, or function, or are in fact nucleoside analogs that
  • 5-Fluorouracil is a clinically important
  • cytoreductive antineoplastic chemotherapy agent that acts in part through incorporation into RNA, producing
  • 5-FU defective RNA; inhibition of thymidylate synthetase by fluorodeoxyuridine monophosphate may also contribute to the cytotoxicity of 5-FU.
  • the clinical utility of 5-FU is limited by its toxicity (especially to bone marrow). Specifically, its clinical utility is limited by a low therapeutic ratio (the ratio of toxic dose to effective dose; a high therapeutic ratio implies that a drug has efficacy with little toxicity).
  • 5-FU and many other chemotherapy agents also affect other tissues, especially gastrointestinal mucosa, producing mucositis, diarrhea and ulceration.
  • Stomatitis (ulceration of mucosa in the mouth), is particularly troublesome to patients, making eating and swallowing painful.
  • Uridine is poorly absorbed after oral administration; diarrhea is dose limiting in humans (van Groeningen et al., Proceedings of the AACR 28:195 [1987]). Consequently, parenteral administration of uridine is necessary for clinically significant reversal of 5-FU toxicity, which requires use of a central venous catheter, since phlebitis has been a problem in early clinical trials when uridine was
  • Orally-active prodrugs of 5FU have been developed which are enzymatically or spontaneously converted to 5FU, generally after absorption from the intestine into the bloodstream. This permits self-administration by patients, without the discomfort of intravenous
  • 5FU prodrugs can in principle provide such sustained availability of 5FU to tumors.
  • 5-Fluoro-1-(tetrahydro-2-furfuryl)uracil is an orally active prodrug of 5-fluorouracil. It is
  • the liver has relatively high levels of the enzyme dihydropyrimidine dehydrogenase, which degrades 5FU, producing metabolites which are not useful in cancer chemotherapy and which furthermore contribute to 5-FU toxicity.
  • the cytotoxicity of 5FU is believed to be a result of its incorporation into nucleotide pools, where certain anabolites exert toxic effects, e.g. 5-fluorodeoxyuridine monophosphate inhibits thymidylate synthetase, thus depriving cells of thymidine for DNA synthesis, and 5-fluorouridine triphosphate incorporation into RNA impairs its normal functions in translation of genetic information.
  • pyrimidine uracil inhibits the catabolism of 5FU without inhibiting its cytotoxicity.
  • the most widely used clinical formulation of FT contains uracil in a 1:4 molar ratio. This permits a significant reduction in the dose of FT required to achieve a
  • uridine, thymidine, thymine, and cytosine are either less effective than uracil or no better in potentiating the antitumor efficacy of FT without unacceptably
  • DHPDHase dihydropyrimidine dehydrogenase
  • FT or 5FU 5-chloro-2,6-dihydroxypyridine
  • CDHP 5-chloro-2,6-dihydroxypyridine
  • this compound also enhances the toxicity of 5FU, so that, in its intended clinical implementation, a third component, oxonic acid, is coadministered to reduce the intestinal toxicity.
  • thymidine when administered at the same time as 5FU increased both the antitumor efficacy of 5FU and its toxicity, so that there was no net increase in
  • pyrimidine nucleosides at doses which are inactive alone, markedly potentiate the antileukemic effects of small doses of FUDR or FU, it has not been possible with any combination to improve significantly and with any degree of regularity the results which can be obtained with maximum tolerated doses of FU or FUDR alone.
  • deoxyuridine by inhibiting the catabolism of the fluoropyrimdines permitted adminstration of lower doses, deoxyuridine there was no improvement in antitumor activity at equitoxic doses of the combination versus FU or FUDR alone.
  • bioavailability after oral administration limit the clinical utility of administration of deoxycytidine, cytidine, and deoxyuridine themselves for modulation of toxicity of chemotherapy agents.
  • Arabinosyl cytosine (Ara-C) is an important agent in the treatment of leukemia, and is also useful as an immunosuppressant. Bone marrow toxicity (myeloid and erythroid) associated with Ara-C administration can be partially prevented by administration of deoxycytidine (Belyanchikova et al. Bull. Exp. Biol. Med. 91:83-85
  • Deoxycytidine also attenuates toxicity of 5-aza-2'- deoxycytidine and arabinosyl 5-azacytosine in cell cultures (K. Bhalla et al. Leukemia 1:814-819 [1987]). Prolonged (5 day) infusion of high doses of deoxycytidine via a central venous catheter was proposed as a means for clinical implementation of modulation of Ara-C toxicity with deoxycytidine (K. Bhalla et al. Leukemia 2:709-710 [1988]).
  • N-phosphonoacetyl-L-aspartic acid is an antineoplastic agent that inhibits the enzyme aspartate transcarbamoylase, an enzyme indirectly involved in biosynthesis of pyrimidine nucleotides. Side effects of PALA primarily involve damage to gastrointestinal
  • Pyrazofurin (a carbon linked pyrimidine analog), 6-azauridine, and 6-azacytidine all interfere with pyrimidine nucleotide synthesis and metabolism.
  • AZT 3'-Azidodeoxythymidine
  • HIV Human Immunodeficiency Virus
  • HIV the infectious agent in AIDS
  • uridine ameliorates AZT-induced toxicity to granulocyte/macrophage progenitor cells without impairing the antiviral actions of AZT (Sommadossi et al., (1988) Antimicrobial Agents and Chemotherapy, 32:997-1001);
  • mice parenteral administration of high doses of uridine provided some amelioration of AZT-induced anemia, but only at uridine doses which increased mortality during the study; a low, non-toxic dose of uridine (500 mg/kg/d) did not reduce AZT-induced
  • progenitor cells in vitro against the cytotoxicity of AZT with preservation of antiretroviral activity progenitor cells in vitro against the cytotoxicity of AZT with preservation of antiretroviral activity.
  • nucleotide precursor orotic acid has antiproliferative effects on human cells, but is especially useful for treating infections with malarial parasites, e.g.,
  • Plasmodium yoelii or Plasmodium falciparum which are dependent on de novo pyrimidine biosynthesis.
  • ddC retroviral infections including HIV
  • side effects of ddC include peripheral neuropathy, mouth ulcers, and reduced platelet counts.
  • the toxicity of ddC on human myeloid progenitor cells in culture can be ameliorated by
  • 438,493 demonstrates the use of acylated derivatives of cytidine and uridine to increase blood cytidine or uridine levels.
  • acyl derivatives of pyrimidine nucleosides have been synthesized for use as protected intermediates in the synthesis of oligonucleotides or nucleoside analogs, e.g. 5'-O-benzoyluridine, triacetylcytidine, and
  • a further object of the invention is to provide compounds and methods to permit administration of higher doses of the chemotherapy agents.
  • a further object of the invention is to provide methods of increasing blood and tissue levels of uridine and cytidine, and their corresponding deoxyribonucieosides deoxycytidine and deoxyuridine through oral administration of a compound or compounds.
  • a further object of the invention is to provide a method for preventing or ameliorating gastrointestinal epithelium damage due to cytotoxic chemotherapy agents.
  • acylated derivatives of non-methylated pyrimidine nucleosides e.g. acylated derivatives of uridine, deoxyuridine, cytidine, or deoxycytidine, which are administered to animals, including mammals such as humans.
  • combination is useful in preventing or ameliorating toxic effects of cytoreductive chemotherapy in animals.
  • the compounds of the invention are useful in the treatment of disorders of hematopoiesis induced by chemical agents; are useful as adjuncts to cancer and antiviral chemotherapy; and are useful for the treatment of other pathological disorders of hematopoiesis induced by chemical agents; are useful as adjuncts to cancer and antiviral chemotherapy; and are useful for the treatment of other pathological disorders of hematopoiesis induced by chemical agents; are useful as adjuncts to cancer and antiviral chemotherapy; and are useful for the treatment of other pathological
  • the compounds useful in attenuating toxicity due to anticancer or antiviral agents have the following general structures:
  • R 1 , R 2 , R 3 and R 4 are the same or different and each is hydrogen or an acyl radical of a metabolite, provided that at least one of said R substituents is not hydrogen, or a pharmaceutically acceptable salt thereof.
  • R 2 , R 2 , and R 3 are the same or different and each is hydrogen or an acyl radical of a metabolite, provided that at least one of said R substituents is not hydrogen, or a pharmaceutically acceptable salt thereof.
  • Compounds of the invention useful in ameliorating toxicity due to anticancer or antiviral chemotherapy agents include the following:
  • R 1 , R 2 , and R 3 are the same, or different, and each is hydrogen or an acyl radical of
  • a an unbranched fatty acid with 5 to 22 carbon atoms.
  • b an amino acid selected from the group consisting of glycine, the L forms of alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine, carnitine and ornithine,
  • a dicarboxylic acid having 3-22 carbon atoms d. a carboxylic acid selected from one or more of the group consisting of glycolic acid, pyruvic acid, lactic acid, enolpyruvic acid, lipoic acid, pantothenic acid, acetoacetic acid, p-aminobenzoic acid, betahydroxybutyric acid, orotic acid, and creatine.
  • R 1 , R 2 , R 3 , and R 4 are the same, or different, and each is hydrogen or an acyl radical of
  • a an unbranched fatty acid with 5 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of phenylalanine, alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine carnitine and ornithine, c. a dicarboxylic acid having 3-22 carbon atoms, d.
  • a carboxylic acid selected from one or more of the group consisting of glycolic acid, pyruvic acid, lactic acid, enolpyruvic acid, lipoic acid, pantothenic acid, acetoacetic acid, p-aminobenzoic acid, betahydroxybutyric acid, orotic acid, and creatine.
  • R 1 , R 2 , and R 3 are the same, or different, and each is hydrogen or an acyl radical derived from
  • a an unbranched fatty acid with 3 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine, carnitine and ornithine,
  • R 1 and/or R 2 may also be acetyl, or a
  • R 1 , R 2 , and R 3 are the same, or different, and each is hydrogen or an acyl radical derived from
  • a an unbranched fatty acid with 3 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine, carnitine and ornithine,
  • R 1 and/or R 2 may also be acetyl, or a
  • hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a
  • hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate.
  • R 1 , R 2 , R 3 or R 4 is a
  • hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a
  • R 1 , R 2 , or R 3 is a
  • hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a
  • hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate.
  • R 1 or R 2 is a hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate.
  • the subject invention relates to the use of acylated derivatives of non-methylated pyrimidine nucleosides, i.e.
  • TAU triacetyluridine
  • the invention also relates to the administration of these pyrimidine nucleoside compounds, alone or in
  • the compounds and methods of the subject invention make it possible to reduce toxicity while maintaining therapeutic efficacy of the antiviral or antineoplastic agent, and
  • the present invention provides compounds and methods for treating or preventing toxic symptoms of antiviral or
  • non-methylated pyrimidine nucleoside as used herein means naturally occurring nucleosides other than thymidine (5-methyldeoxyuridine) or 5-methylcytidine and other similar naturally-occurring methylated nucleosides.
  • non-methylated pyridimidine nucleosides include uridine, cytidine, deoxyuridine, and deoxycytidine.
  • acyl derivative as used herein means a
  • acyl substituents are derived from carboxylic acids which include, but are not limited to, compounds
  • acyl selected from the group consisting of a fatty acid, an amino acid, nicotinic acid, dicarboxylic acids, lactic acid, p-aminobenzoic acid and orotic acid.
  • substituents are carboxylic acids which are normally present in the body, either as dietary constituents or as intermediary metabolites.
  • analog as used herein means a nucleoside chemically modified in either the pyrimidine ring or the ribose (or deoxyribose) moiety by a means other than acylation or attachment of other biologically labile substituents (e.g. phosphorylation of hydroxyl groups on the sugar).
  • nucleoside analogs in the context of this invention, are drugs with structural similarities to the naturally occurring nucleosides, but with antiviral,
  • antineoplastic nucleoside analogs include but are not limited to the following: 5-fluorouracil (5-FU), 5-FU prodrugs (e.g. ftorafur, 5'-deoxyfluorouridine, carmofur), fluorouridine, 2'-deoxyfluorouridine, prodrug derivatives of fluorouridine or 2'-deoxyfluorouridine, fluorocytosine, arabinosyl cytosine, prodrugs of arabinosyl cytosine, cyclocytidine, 5-aza-2'-deoxycytidine, arabinosyl 5-azacytosine, 6-azauridine, azaribine, 6-azacytidine, trifluoro-methyl-2'-deoxyuridine, thymidine, and 3-deazauridine.
  • 5-fluorouracil 5-FU
  • 5-FU prodrugs e.g. ftorafur, 5'-deoxyfluorouridine, carmofur
  • fluorouridine 2
  • nucleoside analogs include but are not limited to the
  • nucleoside analogs are considered to have structural similarities to particular naturally-occurring nucleosides.
  • nucleoside analogs are divided into cytidine analogs if they have an exocyclic amino group in the 4 position of the pyrimidine ring (an amino group in that position signifies the distinction between cytidine and uridine).
  • Nucleoside analogs that are specifically analogs of cytidine include but are not limited to: fluorocytosine, arabinosyl cytosine, prodrugs of
  • nucleoside analogs that are specifically considered to be analogs of uridine include but are not limited to: 5-fluorouracil (5-FU), 5-FU prodrugs (e.g.
  • nucleoside analogs are also specifically analogs of thymidine, e.g. AZT.
  • pharmaceutically acceptable salts means salts with pharmaceutically acceptable acid addition salts of the derivatives, which include, but are not limited to, sulfuric, hydrochloric, or phosphoric acids.
  • pharmaceutically acceptable salts as used herein means that at least two of the compounds of the invention are administered during a time frame wherein the respective periods of
  • hydrocarbylcarbonyl as used herein means an acyl radical of a carboxylic acid in which the atom adjacent to the carbonyl carbon atom is another carbon atom.
  • the parent carboxylic acid may, for example, be a fatty acid, an aromatic acid (e.g. benzoate, nicotinoate, or their congeners), an amino acid, a cycloalkylcarboxylic acid, or a dicarboxylic acid.
  • hydrocarbyloxycarbonyl as used herein means an acyl radical of a carboxylic acid in which the atom adjacent to the carbonyl carbon atom is oxygen which is furthermore covalently linked to another carbon atom. This can also be described as a radical of a carbonate ester of an alcohol, which, when cleaved from a non-methylated pyrimidine
  • nucleoside following administration degrades further into carbon dioxide and an alcohol.
  • Advantageous alcohols are those which are of low toxicity, particularly those which enter readily into normal metabolic or eliminative pathways.
  • fatty acids as used herein means aliphatic carboxylic acids having 2-22 carbon atoms. Such fatty acids may be saturated, partially saturated or polyunsaturated.
  • amino acids as used herein includes, but is not limited to, glycine, the L forms of alanine, valine, leucine, isoleucine, phenylalanine, tyrosine, proline, hydroxyproline, serine, threonine, cysteine, cystine, methionine, tryptophan, aspartic acid, glutamic acid, arginine, lysine, histidine, ornithine, hydroxylysine, carnitine, and other naturally occurring amino acids.
  • dicarboxylic acids as used herein means fatty jcids with a second carboxylic acid substituent.
  • terapéuticaally effective amount refers to that amount which provides therapeutic effects for a given condition and administration regime.
  • the compounds useful in attenuating toxicity due to anticancer or antiviral agents have the following general structures:
  • R 1 , R 2 , R 3 and R 4 are the same or different and each is hydrogen or an acyl radical of a metabolite, provided that at least one of said R substituents is not hydrogen, or a pharmaceutically acceptable salt thereof.
  • R 1 , R 2 , R 3 and R 4 are the same or different and each is hydrogen or an acyl radical of a metabolite, provided that at least one of said R substituents is not hydrogen, or a pharmaceutically acceptable salt thereof.
  • R 1 , R 2 , and R 3 are the same or different and each is hydrogen or an acyl radical of a metabolite, provided that at least one of said R substituents is not hydrogen, or a pharmaceutically acceptable salt thereof.
  • R 1 , R 2 , and R 3 are the same or different and each is hydrogen or an acyl radical of a metabolite, provided that at least one of said R substituents is not hydrogen, or a pharmaceutically acceptable salt thereof.
  • toxicity due to anticancer or antiviral chemotherapy agents include the following:
  • R 1 , R 2 , and R 3 are the same, or different, and each is hydrogen or an acyl radical of
  • a an unbranched fatty acid with 5 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine, carnitine and ornithine,
  • a dicarboxylic acid having 3-22 carbon atoms d. a carboxylic acid selected from one or more of the group consisting of glycolic acid, pyruvic acid, lactic acid, enolpyruvic acid, lipoic acid, pantothenic acid, acetoacetic acid, p-aminobenzoic acid, betahydroxybutyric acid, orotic acid, and creatine.
  • a carboxylic acid selected from one or more of the group consisting of glycolic acid, pyruvic acid, lactic acid, enolpyruvic acid, lipoic acid, pantothenic acid, acetoacetic acid, p-aminobenzoic acid, betahydroxybutyric acid, orotic acid, and creatine.
  • An acyl derivatives of cytidine having the formula:
  • R 1 , R 2 , R 3 , and R 4 are the same, or different, and each is hydrogen or an acyl radical of
  • a an unbranched fatty acid with 5 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of phenylalanine, alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine carnitine and ornithine, c. a dicarboxylic acid having 3-22 carbon atoms, d.
  • a carboxylic acid selected from one or more of the group consisting of glycolic acid, pyruvic acid, lactic acid, enolpyruvic acid, lipoic acid, pantothenic acid, acetoacetic acid, p-aminobenzoic acid, betahydroxybutyric acid, orotic acid, and creatine.
  • a an unbranched fatty acid with 3 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine, carnitine and ornithine,
  • R 1 and/or R 2 may also be acetyl, or a
  • R 1 , R 2 , and R 3 are the same, or different, and each is hydrogen or an acyl radical derived from
  • a an unbranched fatty acid with 3 to 22 carbon atoms
  • b an amino acid selected from the group consisting of glycine, the L forms of alanine, valine, leucine, isoleucine, tyrosine, proline, hydroxyproline, serine, threonine, cystine, cysteine, aspartic acid, glutamic acid, arginine, lysine, histidine, carnitine and ornithine,
  • R 1 and/or R 2 may also be acetyl, or a
  • R 1 , R 2 , or R 3 is a
  • hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a
  • hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate.
  • R 1 , R 2 , R 3 or R 4 is a
  • hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a
  • R 1 , R 2 , or R 3 is a
  • hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a
  • hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate.
  • R 1 or R 2 is a hydrocarbyloxycarbonyl moiety containing 2-26 carbon atoms and the remaining R substituents are independently a hydrocarbyloxycarbonyl or hydrocarbylcarbonyl moiety or H or phosphate. Also encompassed by the invention are the
  • Advantageous compounds of the invention are fatty acid esters of uridine and deoxycytidine, especially those with 4 or fewer carbon atoms in the acyl substituent. Particularly advantageous compounds are fatty acid esters of uridine or deoxycytidine with 2 or 3 carbon atoms in the acyl
  • deoxycytidine particularly those with 3 to 6 carbon atoms in the hydrocarbyloxycarbonyl moiety.
  • prodrugs of the compounds of the invention with enhanced water solubility are prepared by attaching phosphate to a free hydroxyl group on the aldose moiety of the acylated non-methylated pyrimidine nucleoside.
  • compositions of the invention include one or more of the above-noted compounds along with a pharmaceutically acceptable carrier.
  • compositions of the invention include in addition to one or more compounds of the invention and at least one of the following agents which enhance
  • hematopoiesis oxypurine nucleosides, congeners of oxypurine nucleosides, and acyl derivatives of oxypurine nucleosides and their congeners, e.g. fatty acid esters of guanosine or deoxyguanosine (see US Ser . No. 653,882, filed, February 8, 1991, hereby incorporated by reference), a nonionic
  • an interleukin such as IL-1,-2,-3,-4,-5,-6,-7,-8 (advantageously IL-1, 3, or 6)
  • a colony-stimulating factor for example granulocyte colony-stimulating factor (G-CSF), granulocyte/macrophage colony-stimulating factor (GM-CSF), stem cell factor (SCF), erythropoietin (EPO), glucan,
  • G-CSF granulocyte colony-stimulating factor
  • GM-CSF granulocyte/macrophage colony-stimulating factor
  • SCF stem cell factor
  • EPO erythropoietin
  • polyinosine-polycytidine or any other agent having beneficial effects on hematopoiesis.
  • R A H or an acyl radical of a carboxylic acid with 2 to 30 carbon atoms
  • R B H or an acyl radical of a carboxylic acid with 2 to 30 carbon atoms
  • R c H or an acyl radical of a carboxylic acid with 2 to 30 carbon atoms
  • ann L H or OR D
  • R D H or an acyl radical of a carboxylic acid with 2 to 30 carbon atoms
  • R E H or an acyl radical of a carboxylic acid with 2 to 30 carbon atoms, with the proviso that at least one of L and M is H, and
  • Q H, a halogen, NHR F where R F is H or an acyl or alkyl radical containing 1 to 10 carbon atoms, S divalently bound to the carbon in which case the adjacent carbon-nitrogen double bond is a single bond and an H is then attached to that nitrogen, SR G where R G is H or an acyl or alkyl radical
  • R H is H or an acyl or alkyl radical containing 1 to 10 carbon atoms
  • the C-C bond between the 2' and 3' positions of the aldose moiety is optionally present.
  • an acylated nonmethylated pyrimidine nucleoside is formulated with a compound capable of enhancing the uptake and phosphorylation of
  • nucleosides into cells such as insulin or an insulinogenic carbohydrate.
  • the composition comprises at least one compound of the invention and an antiviral or antineoplastic agent (see detailed discussion of these agents in the section below entitled Therapeutic Uses of the Compounds and Compositions of the Invention).
  • the compositions of the invention comprise an acyl derivative of uridine or deoxyuridine and a compound capable of inhibiting uridine phosphorylase.
  • Uridine phosphorylase is the primary enzyme involved in the catabolism of uridine, forming uracil and ribose phosphate.
  • phosphorylase will modify the pharmacokinetics and biological activity of uridine or deoxyuridine produced by deacylation of acylated derivatives of these two non-methylated pyrimidine nucleosides.
  • suitable inhibitors of uridine phosphorylase include but are not limited to 5-benzyl
  • barbiturate or 5-benzylidene barbiturate derivatives including 5-benzyl barbiturate, 5-benzyloxybenzyl barbiturate, 5-benzyloxybenzyl-1-[(1-hydroxy-2-ethoxy)methyl] barbiturate,5-benzyloxybenzylacetyl-1-[(1-hydroxy-2-ethoxy)methyl]barbiturate, and 5-methoxybenzylacetylacyclobarbiturate, 2,2'-anhydro-5-ethyluridine, and acyclouridine compounds, particularly 5-benzyl substituted acyclouridine congeners including but not limited to benzylacyclouridine, benzyloxybenzylacyclouridine, aminomethyl-benzylacyclouridine,
  • the composition comprises an acyl derivative of a non-methylated pyrimidine nucleoside and a compound which inhibits cellular uptake or excretion of non-methylated pyrimidine nucleosides, and thereby promotes maintenance of blood nucleoside levels after enzymatic deacylation of administered doses of acylated derivatives of non-methylated pyrimidine nucleosides.
  • modulators of uridine transport or excretion include but are not limited to dipyridamole, probenicid, lidoflazine or nitrobenzylthioinosine.
  • the composition comprises an acyl derivative of cytidine and a compound capable of inhibiting the enzyme uridine phosphorylase.
  • the composition comprises an acyl derivative of cytidine or deoxycytidine and a compound capable of inhibiting deoxycytidine deaminase.
  • a compound capable of inhibiting deoxycytidine deaminase By inhibiting the deamination of deoxycytidine or cytidine, inhibitors of cytidine deaminase or deoxycytidine deaminase such as tetrahydrouridine or tetrahydro-2'-deoxyuridine modify the efficacy of acyl derivatives of cytidine or deoxycytidine.
  • an inhibitor of cytidine deaminase or deoxycytidine deaminase is used to modify the toxicity of an antiviral or anticancer nucleoside analog (see Example 11).
  • the composition comprises an acyl derivative of a non-methylated pyrimidine nucleoside and an agent or agents with utility in promoting mucosal healing or in reducing discomfort.
  • agents include but are not limited to sucralfate, mixtures of two or more
  • the composition comprises a combination of an acyl derivative of a non-methylated pyrimidine nucleoside and an orally-active
  • antineoplastic nucleoside analog An advantageous combination is an acyl derivative of uridine with an orally active fluorinated pyrimidines, especially prodrugs of 5-fluorouracil.
  • the acyl derivative of a non-methylated pyrimidine nucleoside is mixed with (or otherwise adminstered with) the antineoplastic nucleoside analog in molar ratios, ranging from 1:1 to 12:1. Molar ratios ranging from 2:1 to 8:1 are generally advantageous.
  • Suitable orally-active fluorinated pyrimidines include tegafur, 5'-deoxyfluorouridine, 5-fluorouracil, 5-fluorouridine, 2'-deoxy-5-fluorouridine, N 4 -trimethoxybenzoyl-5'-deoxy-5-fluorocytidine, or acyl
  • compositions are manufactured in the form of a liquid, a suspension, a tablet, a capsule, a dragee, an injectable solution, a topical solution, or a suppository (see discussion of formulation below).
  • a suppository see discussion of formulation below.
  • compounds of the invention are coadministered with the other active agents.
  • the compounds of the invention are useful to prevent or treat damage to the process of hematopoiesis and immune system function in animals.
  • the compounds reduce damage to the process of hematopoiesis by minimizing loss in blood cell counts after bone marrow damage or suppression caused by antiviral or antineoplastic agents which affect nucleotide biosynthesis, metabolism, or utilization.
  • the compounds of the invention are useful in treating humans; however, the invention is not intended to be so limited, it being within the contemplation of the invention to treat all animals that experience a beneficial effect from the administration of the active compounds of the invention.
  • the invention is furthermore embodied in the
  • Specific conditions where advantages are achieved using the compounds, compositions, and methods of the invention include situations where the hematopoietic system has suffered or is likely to suffer damage from chemotherapy, particularly chemotherapy that affects nucleotide biosynthesis, metabolism, or utilization.
  • Such conditions include treating animals, e.g. human patients, subjected to cytoreductive cancer
  • chemotherapy or antiviral chemotherapy are Specifically included are veterinary applications requiring maintenance of blood cell counts.
  • the compounds and compositions are also useful for preventing or treating damage caused by anticancer or
  • the compounds and compositions are optionally administered orally, as a suppository, or parenterally.
  • the compounds and methods of the invention reduce the risk of susceptibility to opportunistic or secondary infections
  • Such agents include hematopoietic growth factors (e.g. G-CSF,
  • GM-CSF GM-CSF, SCF, acylated oxypurine nucleosides and their
  • insulinogenic carbohydrates such as glucose or glucose
  • the white blood cell counts, and particularly the neutrophil counts, of patients treated with standard anti-neoplastic chemotherapy agents e.g., 5-fluorouracil, fluorodeoxyuridine, vinca alkaloids, cyclophosphamide and other nitrogen mustard alkylating agents, daunorubicin, doxorubicin, methotrexate, cytosine arabinoside, 6-mercaptopurine, thioguanosine,
  • standard anti-neoplastic chemotherapy agents e.g., 5-fluorouracil, fluorodeoxyuridine, vinca alkaloids, cyclophosphamide and other nitrogen mustard alkylating agents, daunorubicin, doxorubicin, methotrexate, cytosine arabinoside, 6-mercaptopurine, thioguanosine,
  • podophyllotoxins podophyllotoxins, cisplatin or combinations of such
  • cytoreductive agents are often greatly diminished.
  • daily administration oral or parenteral
  • an effective dose for example, 0.1 - 10.0 grams
  • a compound of the invention such as
  • triacetyluridine (or other acyl derivatives of uridine, cytidine, deoxycytidine, or deoxyuridine) for several days reduces the severity of the neutropenia which typically occurs several days to several weeks after chemotherapy is initiated. This reduces the likelihood of infection throughout the course of treatment, and makes it possible for the patient to receive larger doses of the chemotherapeutic agents and/or to receive repeated doses sooner than comparable patients not treated with the uridine derivative(s). Similarly, chemotherapy-induced alterations in counts of other blood cell types
  • Antineoplastic agents with which the compounds and methods of the invention are particularly useful include: 5-fluorouracil (5-FU), 5-FU prodrugs (e.g. ftorafur, 5'-deoxyfluorouridine, carmofur), fluorouridine, 2'-deoxyfluorouridine, prodrug derivatives of fluorouridine or 2'-deoxyfluorouridine, fluorocytosine (which also has
  • arabinosyl cytosine cyclocytidine, 5-aza-2'-deoxycytidine, arabinosyl 5-azacytosine, N-phosphonoacetyl-L-aspartic acid (PALA), pyrazofurin, 6-azauridine, azaribine, 6-azacytidine, trifluoro-methyl-2'-deoxyuridine, thymidine, and 3-deazauridine.
  • Such antineoplastic agents and various other therapeutic nucleoside analogs act by affecting nucleoside or nucleotide biosynthesis, utilization, or metabolism; hence, amelioration of their toxic effects is accomplished by
  • the compounds of the invention are administered before, during, and/or after administration of the anti-neoplastic or antiviral agents.
  • the compounds of the invention are administered after a dose of a cancer chemotherapy agent, as a means of "rescuing" normal tissues after administration of an effective antineoplastic dose of the agent.
  • Gastrointestinal epithelium is sensitive to cancer chemotherapy agents like fluorouracil. Mucositis, stomatitis, or ulceration of the gastrointestinal mucosa are common side effects of cancer chemotherapy, resulting in discomfort, diarrhea, electrolyte imbalances and weight loss.
  • cancer chemotherapy agents like fluorouracil.
  • Mucositis, stomatitis, or ulceration of the gastrointestinal mucosa are common side effects of cancer chemotherapy, resulting in discomfort, diarrhea, electrolyte imbalances and weight loss.
  • compositions of the invention are useful in preventing or treating damage to the gastrointestinal tract (including the mouth) caused by cancer chemotherapy agents.
  • the compounds and compositions of the invention are optionally administered for this purpose as a solution or suspension in liquid form (as a mouthwash, as a composition to be swallowed, or as an enema), as a capsule, dragee, or tablet, as an injectable solution, or as a suppository.
  • administration of the compounds and compositions of the invention also reduces damage to gastrointestinal mucosa caused by anticancer or antiviral nucleoside analogs.
  • Topical application of the compounds (e.g. to the scalp) of the invention is useful for preventing chemotherapy-induced alopecia.
  • acyl derivatives of uridine are those substituted with short-chain fatty acids, (especially acetate) or with short chain carbyloxycarbonates (e.g. ethoxycarbonate).
  • acyl derivatives of cytidine or deoxyuridine are also useful in treating toxicity due to fluorouracil or related fluorinated pyrimidine analogs.
  • a patient receives a dose of fluorouracil, either as a single treatment agent or as part of a regimen also involving administration of other antineoplastic drugs like methotrexate, leucovorin, PALA, or cyclophosphamide.
  • antineoplastic drugs like methotrexate, leucovorin, PALA, or cyclophosphamide.
  • the patient receives an oral dose of 1 to 10 grams of triacetyluridine.
  • the patient receives additional doses of 5-FU of similar size every 6 to 8 hours over the course of the next 2 to 4 days.
  • the patient may receive additional courses of 5-FU plus TAU on a weekly basis or less frequently.
  • Acyl derivatives of uridine and, secondarily, cytidine are also advantageous for treatment or prevention of toxicity due to N-phosphonoacetyl-L-aspartic acid (PALA), pyrazofurin, 6-azauridine, azaribine, trifluoro-methyl-2'-deoxyuridine, and 3-deazauridine.
  • PKA N-phosphonoacetyl-L-aspartic acid
  • pyrazofurin 6-azauridine
  • azaribine trifluoro-methyl-2'-deoxyuridine
  • 3-deazauridine 3-deazauridine
  • orally-active fluorinated pyrimidine or prodrugs of fluorinated pyrimidines such as 5' deoxy-fluorouridine derivatives like tegafur (5-fluoro-1-(tetrahydro-2-furfuryl)uracil), 5'-deoxyfluorouridine, or related derivatives
  • acyl derivatives of non-methylated pyrimidine nucleosides may be used in several ways.
  • the acyl derivative of a non-methylated pyrimidine nucleoside is administered several hours to one day after a dose of a fluorouracil prodrug such as tegafur, similar to the situation with parenteral administration of fluorouracil described above.
  • a fluorouracil prodrug such as tegafur
  • the delayed administration of the acyl derivative of a non-methylated pyrimidine nucleoside results in reduced toxicity of the fluorinated pyrimidine toward normal tissues.
  • the acyl derivative of a non-methylated pyrimidine nucleoside is administered at the same time as, or within about an hour of, the orally-active antineoplastic agent.
  • Tegafur an orally active 5-fluorouracil prodrug
  • uracil potentiates the antitumor efficacy of 5-fluorouracil produced by degradation of tegafur (during and after absorption from the intestinal tract into the bloodstream) by competing with 5-fluorouracil for the enzyme which breaks down both pyrimidine molecules,
  • Gastrointestinal damage is the primary dose-limiting toxicity of a mixture of tegafur and uracil.
  • Co-administration of tegafur (or other orally active antineoplastic pyrimidine analogs) with an acyl is the primary dose-limiting toxicity of a mixture of tegafur and uracil.
  • acyl derivative of a non-methylated pyrimidine nucleoside and the orally active antineoplastic nucleoside analog are administered in ways and at dosages and molar ratios typically used for administration of uracil and an orally active antineoplastic agent. See, for example, US Patent 4,328,229, hereby incorporated by
  • nucleoside analogs that are specifcally analogs of cytidine, u.g. arabinosyl cytosine or prodrugs thereof, cyclocytidine, 5-aza-2'-deoxycytidine, arabinosyl 5-azacytosine, or 6-azacytidine.
  • advantageous acyl derivatives of deoxycytidine are those substituted with short-chain fatty acids, especially acetate.
  • the antineoplastic agents be used for treating the types of tumors for which they are normally utilized, e.g. Ara-C and its related cytidine analogs are effective in leukemias, fluorouracil and related fluorinated uridine analogs are useful in treating tumors of the colon, stomach, pancreas, and head-and-neck.
  • the antineolastic agents are administered in their normal doses, in which case the compounds of the invention primarily reduce the severity of toxic side effects.
  • the antineoplastic agents are administered in doses higher than normal, in which case the compounds of the invention permit safer administration of such higher, therapeutically aggressive doses of the anticancer drugs.
  • the increases in therapeutic index of anticancer agents resulting from use of the compounds and compositions of the invention permit the use of particular antineoplastic agents for
  • HIV-infected patients especially those whose infection has progressed to "acquired immunodeficiency syndrome" (AIDS)
  • AIDS immunodeficiency syndrome
  • antiviral chemotherapeutic agents such as AZT, which also have detrimental effects on the body's immune function and upon hematopoiesis, further lowering resistance to infections of all kinds.
  • AZT antiviral chemotherapeutic agents
  • invention ⁇ orally, intravenously, or by parenteral injection ⁇ raises the low blood cell counts due to antiviral
  • chemotherapy agents particularly those that modify nucleotide synthesis, metabolism, or utilization, such as AZT or
  • chemotherapeutic treatment of AIDS patients treatment of the patients with these compounds reduces chemotherapeutic side effects (and thus improves the quality of life) and, if appropriate, permits a more intensive chemotherapeutic regimen to be employed.
  • AZT and dideoxycytidine produce deleterious side effects in tissues other than bone marrow, including muscle and the peripheral nervous system.
  • the compounds and compositions of the invention are also useful for treating or preventing such side effects.
  • antiviral nucleoside analogs other than AZT and dideoxycytidine are used to treat viral infections, including but not limited to HIV, herpes, or hepatitis.
  • agents include 5-ethyl-2'-deoxyuridine, 5-iodo-2'-deoxyuridine, 5-bromo-2'-deoxyuridine, 5-methylamino-2'-deoxyuridine, 2 ' ,3'-dideoxycytidin-2'-ene, 3 '-deoxythymidin-2'-ene, 3 '-azido-2',3'-dideoxyuridine,
  • arabinosyluracil dideoxyuridine, 2',3'-dideoxy-3'-fluorothymidine and (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl) cytosine (HPMPC); see also WO 89/09603, hereby incorporated by reference.
  • the compounds of the invention are used to treat or prevent deleterious side effects of these and related other antiviral nucleoside analogs.
  • the compounds and compositions are administered prior to, during and/or after administration of the antiviral agents.
  • Typical antiviral chemotherapy regimens especially for chronic viral infections such as HIV infection, involve daily (often multiple daily) administration of the antiviral agent or agents.
  • the compounds of the invention are administered, several times daily, daily, or less frequently, depending on the clinical effect observed.
  • the antiviral drugs are typically administered in their normal regimens for the types of viral infections for which they are clinically useful. Treatment of patients receiving antiviral nucleoside analogs is undertaken either to reduce side effects of a standard dose or to permit administration of doses of antiviral agents higher than are normally tolerated or
  • acyl derivatives of either or both uridine, cytidine, or deoxycytidine are useful. Particularly advantageous are acyl derivatives of
  • deoxycytidine For oral administration, acyl derivatives of deoxycytidine, uridine, and cytidine substituted with short chain fatty acids (particularly acetate) or with short chain carbyloxycarbonates (e.g. ethoxycarbonate), are advantageous.
  • a patient receives AZT two to four times daily, and must generally do so
  • Malarial parasites e.g. Plasmodium yoelii or Plasmodium falciparum
  • Plasmodium yoelii or Plasmodium falciparum are dependent upon de novo synthesis pathways for pyrimidine nucleotide biosynthesis; mammalian cells in general can utilize either de novo pathways or "salvage" pathways, tnrough which advanced nucleotide precursors such as uridine or cytidine are incorporated into intracellular nucleotide pools.
  • 5-Fluoroorotate an analog of the pyrimidine nucleotide precursor orotic acid, is toxic toward malarial parasites which are dependent on de novo pyrimidine biosynthesis.
  • Other inhibitors of de novo pyrimidine biosynthesis such as PALA, pyrazofurin or 6-azauridine are also similarly toxic toward malaria parasites.
  • Inhibitors of pyrimidine biosynthesis including especially fluoroorotate, are also toxic toward mammals.
  • Such agents include the acyl derivatives of uridine or cytidine of the invention.
  • an effective anti-malarial dose of fluoroorotate is administered.
  • an acyl derivative of uridine or cytidine is particularly advantageous is administered, in a dose
  • acylated uridine or cytidine derivative such as
  • triacetyl uridine range from 1 to 10 grams, administered as often as needed to minimized fluoroorotate toxicity, e.g. one to four times per day. Doses of fluoroorotate or uridine are optionally repeated as necessary to overcome the malarial infection and to reduce host toxicity respectively.
  • compositions of the invention are administered orally, by parenteral injection, intravenously, topically, or by other means, depending on the condition being treated.
  • triacetyluridine or other acyl derivatives of uridine, cytidine, deoxycytidine or deoxyuridine
  • uridine or other acyl derivatives of uridine, cytidine, deoxycytidine or deoxyuridine
  • the compounds and compositions of the invention are administered chronically or intermittently.
  • the compounds and compositions are administered prior to, during, or after an exposure to cytoreductive or antiviral chemotherapy agents, depending on the characteristics of the toxicity of the chemotherapy agents.
  • acyl derivatives of uridine, cytidine, deoxycytidine, or deoxyuridine for oral administration are those substituted with short chain (2-6 carbon) fatty acids on the hydroxyl groups of their ribose or deoxyribose rings.
  • hydrocarbyloxycarbonyl radicals containing 3-7 carbon atoms hydrocarbyloxycarbonyl radicals containing 3-7 carbon atoms.
  • Dosages for orally adminstered acyl derivatives of uridine, cytidine, deoxycytidine or deoxyuridine typically range from 0.5 to 20 grams per day, most commonly 2 to 10 grams per day.
  • deoxycytidine or deoxyuridine are administered orally in capsule or tablet form, although solutions, emulsions, or suspensions are also useful for oral administration.
  • the compounds of the invention are optionally formulated in biodegradable, bioerodible, or other gradual-release matrices for sustained release of the compounds after oral administration or subcutaneous implantation.
  • the compounds are optionally formulated in liposomes.
  • pharmacologically active compounds optionally are combined with suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate delivery of the pharmacologically active compounds.
  • compositions are administered, for example, orally, rectally, vaginally, or released through the buccal pouch of the mouth, and are optionally applied in solution form by injection, orally or by topical administration.
  • the compositions may contain from about 0.1 to 99 percent, preferably from about 50 to 90 percent, of the active compound (s), together with the excipient (s).
  • the active compounds are suspended or dissolved in aqueous medium such as sterile water or saline solution, l.ijectable solutions or suspensions optionally contain a surfactant agent such as polyoxyethylenesorbitan esters, sorbitan esters, polyoxyethylene ethers, or solubilizing agents like propylene glycol or ethanol.
  • aqueous medium such as sterile water or saline solution
  • l.ijectable solutions or suspensions optionally contain a surfactant agent such as polyoxyethylenesorbitan esters, sorbitan esters, polyoxyethylene ethers, or solubilizing agents like propylene glycol or ethanol.
  • the active compounds typically contains 0.01 to 5% of the active compounds.
  • the active compounds optionally are dissolved in pharmaceutical grade vegetable oil for intramuscular injection.
  • preparations contain about 1% to 50% of the active compound(s) in oil.
  • Suitable excipients include fillers such as sugars, for example lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch or potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethyl cellulose and/or polyvinyl pyrrolidone.
  • fillers such as sugars, for example lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch or potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethyl
  • Auxiliaries include flow-regulating agents and
  • lubricants for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate and/or polyethylene glycol.
  • Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices.
  • suitable coatings which, if desired, are resistant to gastric juices.
  • concentrated sugar solutions are used, which optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate are used.
  • Dyestuffs or pigments are optionally added to the tablets or dragee coatings, for example, for identification or in order to characterize different compound doses.
  • compositions of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes.
  • pharmaceutical preparations for oral use are obtained by combining the active compound(s) with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
  • compositions which are useful for oral delivery include push-fit capsules made of gelatin, as well as soft-sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules contain the active compound(s) in the form of granules which
  • the active compounds are preferably dissolved or suspended in suitable liquids such as fatty oils, liquid paraffin, or polyethylene glycols.
  • suitable liquids such as fatty oils, liquid paraffin, or polyethylene glycols.
  • stabilizers optionally are added.
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water soluble form, for example, water soluble salts.
  • & ⁇ spensions of the active compounds as appropriate in oily injection suspensions are administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or tri-glycerides.
  • Aqueous injection suspensions optionally include substances which increase the viscosity of the
  • suspension optionally contains stabilizers.
  • the active compounds are N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidedoxedoxifenoethyl-N-oxidethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidethyl
  • Suitable lipophilic solvents or vehicles include fatty oils, for example sesame oil or coconut oil, or synthetic fatty acid esters, for example ethyl oleate or triglycerides.
  • the active compounds are N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidedoxedoxifenoethyl-N-oxidethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidethyl
  • formulated in vehicles suitable for direct treatment of gastrointestinal mucosa examples include mouthwashes, liquids (solutions or suspensions) to be swallowed, or viscous fluids (e.g. solutions of methylcellulose,
  • rectally especially for treatment of the colon and rectum, include, for example, suppositories which consist of a
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base are useful.
  • Base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • nucleosides are synthesized by reacting a pyrimidine
  • An activated carboxylic acid is one that has been treated with appropriate reagents to render its carboxylate carbon more susceptible to nucleophilic attack than is the case in the original
  • carboxylic acid examples include acid chlorides, acid anhydrides, n-hydroxysuccinimide esters, or carboxylic acids activated with BOP-DC.
  • Carboxylic acids are alternatively linked to pyrimidine nucleosides with coupling reagents like dicyclohexylcarbodiimide (DCC).
  • acyl compounds of the invention when the acid source of the desired acyl derivative has groups which interfere with the acylation reactions, e.g., hydroxyl or amino groups, these groups are blocked with pro tecting groups, e.g., t-butyldimethylsilyl ethers or t-BOC groups, respectively, before preparation of the anhydride.
  • pro tecting groups e.g., t-butyldimethylsilyl ethers or t-BOC groups
  • lactic acid is converted to 2-t-butyldimethylsiloxypropionic acid with t-butyl- dimethylchlorosilane, followed by hydrolysis of the resulting silyl ester with aqueous base.
  • the anhydride is formed by reacting the
  • the N-t-BOC or N-CBZ derivative is prepared, using standard techniques, which is then converted to the anhydride with DCC.
  • acids containing more than one carboxylate group e.g., succinic, fumaric, or adipic acid
  • the acid anhydride of the desired dicarboxylic acid is reacted with a pyrimidine nucleoside in pyridine or pyridine plus dimethylformamide or
  • Amino acids are coupled to the exocyclic amino groups of cytosine and deoxycytosine, and to hydroxyl groups on the aldose moiety of pyrimidine nucleosides, by standard methods using DCC in a suitable solvent, particularly a mixture of (i) methylene chloride and (ii) dimethylacetamide or
  • pyrimidine nucleosides are prepared by reacting the nucleoside with the appropriate carbylchloroformate in a solvent such as pyridine or pyridine plus dimethylformamide under anhydrous conditions. The solvent is removed under vacuum, and the residue is purified by column chromatography. It will be obvious to the person skilled in the art that other methods of synthesis may be used to prepare the
  • Example 1 Oral administration of triacetyluridine ameliorates hematologic toxicity of 5-fluorouracil
  • mice Forty-five female Balb/C mice (20 grams each) were given 5-fluorouracil (150 mg/kg, i.p.) at 12:00 noon on the initial day of the experiment. These animals were then divided into 5 groups: control (water, p.o.), oral uridine at 400 mg/kg/dose, oral uridine at 800 mg/kg/dose, parenteral (i.p.) uridine at 400 mg/kg/dose, and oral TAU at 500 mg/kg/dose.
  • control water, p.o.
  • oral uridine 400 mg/kg/dose
  • oral uridine 800 mg/kg/dose
  • parenteral (i.p.) uridine at 400 mg/kg/dose
  • oral TAU at 500 mg/kg/dose.
  • Groups received their designated treatment at 2:00 p.m., 4:00 p.m., and 6:00 p.m. on the day of 5-FU administration, and at 9 a.m., 11:00 a.m., 1:00 p.m., 3:00 p.m. and 5:00 p.m. on the following day.
  • Each dose of uridine or TAU was administered in 0.2 ml of water by gavage or in 0.2 ml of saline by i.p. injection, as appropriate.
  • Seven days after administration of 5-FU blood (0.2 - 0.3 ml) from five mice in each group was collected from the suborbital sinus into EDTA for subsequent differential blood cell counting.
  • mice were sacrificed by cervical dislocation; femurs were removed, and their cell contents were expelled for counting; spleens were also removed and weighed. Thirteen days after administration of 5-FU, the remaining four mice in each group were bled, sacrificed, and their spleens removed.
  • 5-FU administration resulted in declines in counts of all blood cell types examined. Seven days after administration of 5-FU, neutrophil, lymphocyte, and platelet counts in animals treated with oral TAU were significantly higher than in control animals (Table 1). It is particularly noteworthy that leukocyte counts in the mice that received TAU were higher than in mice that received an equimolar dose of uridine by intraperitoneal injection. Cell counts in the mice that received oral TAU were also higher than in mice that received uridine orally at either equimolar (400 mg/kg/dose) or twice equimolar doses (800 mg/kg/dose).
  • Platelet counts were within the normal range (700-800 K/ ⁇ l) in the mice that received oral TAU and i.p. uridine. They were subnormal in the other groups, and lowest in the control mice (Table 1). Bone marrow cell counts were significantly greater in the mice treated with TAU orally and with uridine parenterally than any of the other groups (Table 2)
  • neutrophil and RBC levels were higher in the group that received TAU than in the other treatment groups, including mice that received an equimolar dose of uridine by intraperitoneal injection (Table 3).
  • Spleen weight is an index of hematopoietic activity in mice recovering from bone marrow damage. Eleven days after 5-FU, spleen weight was significantly higher in mice given oral TAU compared to the other treatment groups; spleens were smallest in the control group (Table 4).
  • TAU 500 oral 5.9 ⁇ .4* 1.20 ⁇ .23** 4.72 ⁇ .36** 723 ⁇ 57 ** 8.27 ⁇ .16
  • Example 2 TAU accelerates hematopoietic recovery in 5-FU-treated animals in a dose dependent manner
  • test compounds were then given at 3:00, 5:00, 7:30, and 10:00 p.m. on the day of 5-FU administration; at 9:00 a.m., 11:00 a.m., and 1:00, 3:00, 6:00, and 10:00 p.m. the following day; and a final administration at 11:00 a.m. two days following the single 5-FU injection.
  • Each treatment was given orally in a volume of 0.2 ml water (by gavage), except the highest dose of TAU which was administered in 0.4 ml of water.
  • lymphocyte counts were significantly elevated compared to those in the cjntrol group.
  • Red blood cell counts were significantly increased in the TAU 500 group (8.72 ⁇ 0.18 ⁇ 10 6 per microliter) and the TAU 1000 group (8.63 ⁇ 0.16 ⁇ 10 6 per microliter) compared to controls (7.90 ⁇ 0.09 ⁇ 10 6 per microliter).
  • the hematocrit followed a similar pattern (Table 7).
  • Table 7 Effect of increasing doses of TAU on hematocrit (HCT) in mice seven (7d) and eleven (lid) days after 5-FU administration.
  • Example 3 Acyl derivatives of uridine ameliorate bone marrow toxicity of 5-fluorouracil
  • mice Ninety-eight female Balb/C mice weighing approximately 20 grams each were given a one-time 150 mg/kg injection (i.p.) of 5-FU at 1 p.m. on the initial day of the study. These animals were then divided into seven groups: control (saline), vridine (300 mg/kg/treatment), triacetyluridine (TAU; 455 mg/kg/treatment), benzoyluridine (BU; 428 mg/kg/treatment), ethoxycarbonyl (ECU; 389 mg/kg/treatment), octanoyluridine (OU; 455 mg/kg/treatment), and valeryluridine (VU; 403
  • TAU increased bone marrow cellularity by 40% over control values (4.50 ⁇ .77 ⁇ 10 3 per microliter versus 2.78 ⁇ .45 ⁇ 10 3 per microliter, respectively).
  • Platelet counts were significantly greater in the groups treated with uridine (785.3 ⁇ 57.5 ⁇ 10 3 per microliter; p ⁇ .02), BU (829.6 ⁇ ⁇ 10 3 per microliter; p ⁇ .01),. and VU (825.7 ⁇ 26.7 ⁇ 10 3 per microliter; p ⁇ .002) than those in the saline-treated controls (523.2 ⁇ 71.4 ⁇ 10 3 per microliter).
  • octanoyluridine having the longest carbon chain of any of the other derivatives, proved to be somewhat detrimental. There were not enough animals from this group to provide day 11 data. However, in dose optimization studies (see Example 3A), octanoyluridine administered at a lower dose showed very beneficial effects on hematopoietic recovery following 5-FU.
  • WBC White blood cell counts
  • Neut neutrophil counts
  • Lym lymphocyte counts
  • K/ ⁇ l red blood cell counts
  • mice Fourteen Balb/C female mice weighing approximately 20 grams each were given a one-time 75 mg/kg i.p. injection of 5-FU at 11:00 a.m. on the initial day of the study. Half of these animals were subsequently treated with Oct-U (100 mg/kg/treatment, i.p.), while the other half (controls) were injected with physiological saline. Administration of Oct-U and saline occurred at 2:30, 4:30, and 7:00 p.m. on the initial day, and at 9:30 a.m., 12:00 noon, 2:30, and 5:00 p.m the following day. An additional group of seven mice (basals) received no 5-FU and no treatments.
  • mice receiving 5-FU with octanoyl uridine ameliorates the toxic effects of 5-FU on hematopoiesis.
  • Example 4 Plasma uridine levels after administration of acyl derivatives of uridine
  • Plasma uridine levels were determined in mice at various times (15 minutes, 30 minutes, 1 hour, and 2 hours) after administration of the acyl derivatives of uridine utilized in
  • BU ethoxycarbonyluridine
  • ECU ethoxycarbonyluridine
  • OU octanoyluridine
  • VU valeryluridine
  • the doses of the acyl derivatives of uridine are the molar equivalent of 300 mg/kg uridine.
  • blood samples 200 ⁇ l were taken from mice via the retro-orbital sinus and immediately centrifuged.
  • 75 ⁇ l of the resulting plasma was deproteinized with 2 volumes of methanol followed by centrifugation.
  • the supernatant was lyophilized and reconstituted with 50 mM potassium phosphate buffer, pH 6.0, and analyzed for uridine content by HPLC on a reverse phase (C 18 ) column.
  • Uridine was separated from other plasma
  • Uridine was detected and quantified by UV absorbance at 260 nM.
  • Plasma uridine levels in control animals were 1.1 ⁇ 0.1 ⁇ M.
  • acylated uridine derivatives provide sustained formation of uridine via gradual deacylation. This may not be reflected in plasma uridine levels, since cellular uptake of uridine can remove uridine from the circulation as it is formed by deacylation of the acylated uridine derivatives. It is important to note that the acylated uridine derivatives are generally superior to an equimolar quantity of uridine in attenuating toxicity due to 5-FU (Table 8 in Example 3)
  • the purpose of this experiment was to assess and compare the ability of uridine and TAU to increase the therapeutic index of 5-FU in a tumor-bearing mouse model.
  • CD8F1 (BALB/C ⁇ DBA/8) female mice with first generation transplants of CD8F1 spontaneous mammary
  • adenocarcinoma were treated with a weekly chemotherapy regimen which included a single dose of 5-FU (150 mg/kg) followed by various rescue strategies.
  • the average tumor size was 157 mg at the start of the chemotherapy.
  • the weekly chemotherapy course was completed three times.
  • 5-FU alone 5-FU (150 mg/kg i.p.)
  • 5-FU + vehicle 5-FU (150 mg/kg i.p.)
  • Uridine (3,500 mg/kg i.p.) 5.
  • 5-FU + oral uridine 5-FU (150 mg/kg i.p.) +
  • Uridine (5,000 mg/kg p.o.) 2 6.
  • 5-FU + oral TAU 5-FU (150 mg/kg i.p.) + TAU
  • TAU and uridine are effective in rescuing tumor-bearing mice from the toxic effects of 5-FU. Both agents increase the therapeutic index of 5-FU in tumor-bearing mice, and allow higher doses of the drug to be tolerated, with a commensurate increase in anti-cancer effect.
  • the purpose of this experiment was to assess and compare the ability of TAU and uridine to increase the therapeutic index of 5-FU when used in combination with Phosphonacetyl-L-aspartate (PALA), methotrexate (MTX), and leucovorin (LV), in a drug dosing regimen which increases the cytotoxic potential of 5-FU.
  • Phosphonacetyl-L-aspartate Phosphonacetyl-L-aspartate
  • MTX methotrexate
  • LV leucovorin
  • mice were divided into four groups of ten animals each. 1.
  • Control Saline
  • Uridine i.p.: Uridine (3,500 mg/kg)
  • Uridine oral
  • Uridine 4,4,000 mg/kg
  • TAU oral TAU (6,066 mg/kg 1 )
  • TAU and uridine improve the therapeutic index of 5-FU used in this clinically relevant combination of agents.
  • Oral TAU was as effective as intraperitoneally-administered uridine and more effective than an equimolar dose of orally-administered uridine.
  • Example 7 Oral administration of diacetyldeoxycytidine attenuates hematopoietic toxicity of arabinosyl cytosine
  • DADC diacetyldeoxycytidine
  • PdC palmitoyldeoxycytidine
  • chemotherapeutic agent arabinosylcytosine (Ara-C) on the hematopoietic system chemotherapeutic agent arabinosylcytosine (Ara-C) on the hematopoietic system.
  • mice Twenty-one Balb/C female mice weighing approximately 20 grams each received a daily intraperitoneal injection of Ara-C (100 mg/kg) for five days. These mice were divided into three treatment groups: oral administration of water (controls); oral administration of DAdC (411 mg/kg/treatment); and
  • mice intraperitoneal administration of PdC (200 mg/kg/treatment in 0.2% Tween 80). Mice were treated with either water, DAdC, or PdC twice daily, at 9 a.m. and 6 p.m. Treatment volume in each case was 0.2 ml. An additional seven mice received no Ara-C and no treatment at all (basals).
  • Ara-C administration resulted in significantly depressed spleen weights, WBC counts, total neutrophil counts,
  • lymphocyte counts, and platelet counts in the control mice compared to the basal mice were compared to the basal mice. No toxic effects of Ara-C were observed on erythropoiesis per se (RBC counts, hemoglobin, and hematocrit).
  • Spleen weight is in milligrams
  • WBC neutrophil
  • platelet counts are expressed as K/ ⁇ l.
  • Example 8 Orally administered TAU ameliorates the
  • TAU triacetyluridine
  • chemotherapeutic agent azidothymidine (AZT).
  • AZT Basal (no AZT, no treatment), control (AZT, water), and TAU (AZT, TAU at 460 mg/kg/treatment).
  • AZT was administered ad libitum in the drinking water at a concentration of 1.5 mg/ml throughout the course of the experiment.
  • the volume of AZT solution consumed in all treatment groups was similar and averaged 2.25 ml per day per mouse, resulting in a daily AZT dose of about 170 mg/kg per day.
  • Water and TAU were administered in a volume of 0.2 mL three times per day for the first 24 days of the study, and twice each day thereafter.
  • mice All of the animals were weighed on the day the experiment began, once each week, and immediately prior to sacrifice. After weighing the mice on days 24 and 35, blood (0.2 - 0.3 ml) was collected from seven mice in each group by retro-orbital bleeding into EDTA for subsequent differential blood cell counting, including reticulocytes. Mice were then sacrificed by cervical dislocation; their right femurs removed and the contents expelled for cell counting; and their spleens removed and weighed.
  • mice treated with TAU (19.51 ⁇ 0.38) were nearly identical to those of basals and significantly greater (p ⁇ .005) than those of controls at this same time point. This trend was also observed on day 13, although there were no statistically significant differences between groups.
  • the body weights of the AZT controls were statistically depressed compared to both basals (p ⁇ .001) and TAU-treated mice (p ⁇ .05). At this time point the body weights of the TAU-treated animals were also less than those of basal animals.
  • the RBC counts of the mice receiving AZT only (8.49 ⁇ 0.16) were significantly reduced (p ⁇ .01) compared to basals (9.07 ⁇ 0.11).
  • the RBC counts of those animals treated with TAU (9.01 ⁇ 0.09) were not significantly different from basals and were significantly greater (p ⁇ .02) than those of
  • mice treated with TAU had reticulocyte levels significantly higher (0.371 ⁇ 10° per microliter) than both basals (p ⁇ .001) and controls (p ⁇ .01).
  • TAU ameliorates the detrimental effects of AZT on erythropoiesis in mice.
  • Red blood cell (RBC) counts are in
  • HCT hematocrit
  • HGB hemoglobin
  • TAU attenuates AZT-induced cell damage.
  • Platelet counts (PLT) are in K/ ⁇ l, mean cell volume (MCV) is in fl, and mean cell hematocrit (MCH) is measured in picograms.
  • the purpose of this experiment was to test the efficacy of orally administered TAU in reversing the hematopoietic damage caused by parenteral administration of azidothymidine (AZT).
  • AZT 100 mg/kg i.p.
  • mice were given three times daily at 9 a.m., 4 p.m., and at 10 p.m. to fifty-six female Balb/C mice weighing approximately 19 grams each.
  • Treatment volume was 0.2 ml for the control and TAU 460 groups, 0.1 ml for the TAU 230 group, and 0.4 ml for the TAU 920 group.
  • One additional group of fourteen mice was not given AZT or any treatments (basal) .
  • Bone marrow cellularity was significantly greater (p ⁇ .05) in the group treated with the 230 mg/kg TAU (8.89 ⁇ 0.46) than in the group receiving AZT alone (7.54 ⁇ 0.23).
  • AZT administration resulted in a drop in RBC levels from 9.14 ⁇ 0.10 ⁇ 10 6 per microliter (basals) to 8.80 ⁇ 0.31 ⁇ 10 6 per microliter (controls). No such decrement was seen in mice receiving TAU (460 mg/kg/treatment) and AZT (9.15 ⁇ 0.07 ⁇ 10 6 per microliter).
  • mice given AZT alone for 13 days showed statistically significant evidence of hematopoietic damage.
  • WBC counts, RBC counts, hemoglobin, hematocrit, reticulocyte counts, total neutrophil counts, and lymphocyte counts were all significantly depressed, while mean cell hematocrit and platelet counts were significantly elevated.
  • Concomitant treatment of mice receiving AZT with TAU resulted in statistically significant improvement in each and all of these measures (Table 17 and 18).
  • Example 10 Oral administration of diacetyldeoxycytidine (DAdC) ameliorates hematopoietic toxicity produced by DADC
  • AZT 100 mg/kg i.p.
  • AZT 100 mg/kg i.p.
  • mice were given three times daily at 9 a.m., 4 p.m., and at 10 p.m. to twenty-eight female DBA mice weighing approximately 20 grams each.
  • Treatment volume was 0.2 ml.
  • One additional group of fourteen mice was not given AZT or any treatment (basal) .
  • mice During the first few days of treatment a total of eight mice died from accidents occurring during oral administration of the water and DAdC. Therefore, the number of animals in the control group and the DAdC group were reduced on the days of sacrifice as follows: On day 6 there were four mice in the control group and five in the DAdC group; on day 13 there were seven in the control group and three in the DAdC group. The number of basal animals was seven at both time points.
  • mice collected from mice in each group by retro-orbital bleeding into EDTA for subsequent differential blood cell counting, including reticulocytes. The animals were then sacrificed by cervical dislocation; their right femurs removed and the contents expelled for cell counting; and their spleens removed and weighed.
  • HCT HCT
  • reticulocyte counts Table 17
  • Platelet counts were significantly elevated in these control mice. Bone marrow cell counts were also reduced 23% in the control group compared to the basals.
  • mice receiving AZT but also treated with DAdC had only a slight (2.5%) reduction in bone marrow cellularity and were not statistically different from, basals.
  • RBC counts, HGB, HCT, and reticulocyte counts in the DAdC group were not different from those in the basal group, but were significantly greater than those in the control group (Table 19).
  • mice given AZT alone for 13 days showed statistically significant evidence of hematopoietic damage in nearly every category compared to basal animals.
  • Concomitant treatment of mice with DAdC markedly attenuated or reversed the AZT-induced erythropoietic damage (Table 20) as was seen on day 6.
  • mice receiving AZT with DAdC significantly improves hematopoietic function, especially erythropoiesis.
  • Example 11 Orally administered diacetyldeoxycytidine (DAdC) or tetrahydrouridine (THU) ameliorate the detrimental effects on the hematopoietic system of intraperitoneally administered AZT in Balb/C mice
  • the purpose of this experiment was to test the efficacy of orally administered DAdC or parenterally administered THU in reversing the hematopoietic damage caused by parenteral administration of the antiviral chemotherapeutic agent azidothymidine (AZT).
  • AZT 100 mg/kg i.p.
  • AZT 100 mg/kg i.p.
  • mice weighing approximately 20 grams each.
  • Treatment volume was 0.2 ml.
  • One group of seven mice was not given AZT or any treatment (basal).
  • mice in each group On day thirteen blood (0.2 - 0.3 ml) was collected from mice in each group by retro-orbital bleeding into EDTA for subsequent differential blood cell counting, including reticulocytes. The animals were then sacrificed by cervical dislocation; their right femurs removed and the contents expelled for cell counting; and their spleens removed and weighed. Results
  • Example 12 Plasma uridine levels after oral administration of uridine or triacetyluridine (TAU), with or without
  • TAU is a more effective orally-active agent for elevating plasma uridine levels than is uridine itself, and furthermore to demonstrate the effect of dipyridamole (DPM), a nucleoside uptake-blocker which also has antiviral activity, on plasma uridine levels after administration of TAU or uridine.
  • DPM dipyridamole
  • mice with a body weight of 20 grams were divided into four groups of 8 animals each: 1. Uridine (1000 mg/kg) p.o.
  • Uridine was administered orally by gavage as an aqueous solution in a volume of 0.4 ml.
  • TAU was administered by gavage in an emulsion vehicle (1:1 corn oil/water with 2.5% Tween 80).
  • mice from each group were bled from the suborbital plexus at each time point: 0 (Basal uridine levels before TAU or uridine administration), 0.5, 1, 2, and 4 hours after administration of TAU or uridine
  • Plasma samples (0.1 ml) were deproteinized by addition of 0.2 ml methanol followed by centrifugation. Samples were lyophilized and then reconstituted with HPLC buffer (100 mM ammonium acetate, pH 6.5, for subsequent assay of uridine by reverse phase HPLC with UV absorbance detection (254 nm).
  • HPLC buffer 100 mM ammonium acetate, pH 6.5
  • Dipyridamole further enhanced (approximately two-fold) the amplitude (peak uridine levels of 460 micromolar) and duration of blood uridine levels after oral administration of TAU.
  • Dipyridamole similarly improved blood uridine level maintenance after oral uridine, although levels were much lower than in the corresponding mice that received TAU (peak plasma uridine levels of 20 micromolar).
  • Plasma uridine levels ( ⁇ M) after TAU or uridine administration Plasma uridine levels ( ⁇ M) after TAU or uridine administration
  • Plasma uridine levels after oral administration of TAU were much higher than were observed after oral administration of uridine. TAU is thus a much better source of plasma uridine after oral administration than is uridine itself.
  • Example 13 Modulation of toxicity of oral tegafur with TAU and uracil
  • Tegafur 5-fluoro-1-(tetrahydro-2-furfuryl)uracil
  • 5-fluorouracil 5-fluoro-1-(tetrahydro-2-furfuryl)uracil
  • tegafur is typically administered orally in a
  • tegafur formulated with uracil is currently used clinically in humans.
  • Uracil potentiates the activity of the 5-FU formed from tegafur by competitively inhibiting dihydrouracil dehydrogenase, an enzyme which degrades 5-FU.
  • mice After administration of high doses of tegafur+uracil, mice lose a substantial amount of body weight, indicating gastrointestinal toxicity. After oral administration, it is believed that uracil potentiates the local toxicity of 5-FU formed in intestinal cells during passage of Tegafur into the bloodstream. It is therefore desirable to utilize with tegafur, or other orally active prodrugs of 5-FU, an agent which inhibits breakdown of 5-FU (or otherwise potentiates its activity) primarily in the circulation (after absorption) rather than locally in the gut.
  • Triacetyluridine like other acyl derivatives of uridine and cytidine of the invention, is converted to uridine and uracil during and after absorption into the bloodstream; when present at the same time as 5-FU, both uridine and uracil are capable of potentiating 5-FU cytotoxicity. Therefore, the cytotoxicity of oral tegafur+uracil versus tegafur+TAU was assessed. Blood cell counts were utilized as an index of cytotoxicity of 5-FU in the circulation (and by extension, of antitumor potency). Body weight loss was used as an index of gastrointestinal toxicity.
  • mice received tegafur by oral intubation in a dose of 400 mg/kg per mouse.
  • the initial body weight of mice in each group was 19.010.6 grams.
  • One of these groups also received uracil in a molar ratio of 4:1 to the tegafur dose, and another group received TAU, also in a 4:1 molar ratio to tegafur. Body weights were monitored. Six days after tegafur administration, blood samples were taken for differential cell counts.
  • Tegafur plus uracil produced a more severe drop in neutrophil counts than did tegafur alone, and also reduced platelet counts and caused a substantial (29%) loss of body weight.
  • Tegafur plus TAU produced blood cell count changes similar to those found after tegafur plus uracil, but did not cause a change in body weight.
  • the blood cell counts found after oral administration of either tegafur+TAU or tegafur+ uracil are similar to those observed after a therapeutically effective dose of 5-fluorouracil (e.g. 150 mg/kg) administered
  • tegafur+uracil is unacceptable in the context of cancer chemotherapy.
  • the excellent systemic cytotoxicity of oral tegafur+TAU (better than tegafur alone and at least equivalent to tegafur+uracil) and the absence of a loss in body weight (especially in contrast to the marked weight loss in animals receiving tegafur+TAU) indicate that TAU is useful in
  • Combination of an orally active fluorouracil prodrug with an acylated non-methylated pyrimidine nucleoside derivative therefore permits better oral delivery of therapeutically effective amounts of the important antineoplastic drug fluorouracil than is obtained with current methods and compositions.
  • Example 14 Antitumor efficacy of tegafur and TAU vs tegafur and uracil
  • Tegafur is an orally active prodrug of fluorouracil
  • FT-uracil combination is damage to the intestinal mucosa. Increasing doses of FT-uracil also result in hematopoietic damage.
  • the purpose of the present study was to compare the antitumor efficacy of FT in combination with either uracil or triacetyluridine (TAU) in rats bearing the Walker 256
  • FT + TAU inhibits tumor growth as well as FT + uracil, while causing less intestinal toxicity (body weight changes). Blood cell damage was also assessed and compared.
  • FT was administered at doses of 40, 60, and 80 mg/kg/day in a 1:4 molar ratio with either uracil or TAU.
  • the vehicle used was 1% hydroxypropylmethylcellulose.
  • Treatment was initiated (day 1) five days after tumor implantation. Vehicle and vehicle plus drugs were administered orally by gavage each day for seven days (day 1 through 7) in a volume of 1.2 milliliters per 100 grams of body weight. Tumor size was measured in situ on day 8. On day 10 blood samples were obtained and, after sacrifice of the animals, tumor size and weight were determined.
  • Body weights were determined prior to treatment on days 1, 3, 5, and 7. On day 10 the animals were weighed prior to sampling and sacrifice. Body weights are expressed as the percent of body weight change over the course of the
  • Tumor size was measured in situ on day 8 and the tumor volume was then calculated using the following formula: length ⁇ width 2
  • T/C % is: mean tumor size in drug-treated rats ⁇ 100 mean tumor size in control rats
  • FT + uracil and FT + TAU have greater anti-tumor efficacy and preserves body weight better than equivalent doses of FT + uracil at each dose of FT.
  • FT 60 the tumor volume and T/C percent for the FT-TAU treated group are less than half those of the uracil-treated group.
  • the body weight change is 52.1% in the FT-TAU group compared to 7.8% in the FT-uracil group. Higher doses of FT are more effective in preventing tumor growth than lower doses.
  • Tumor and body weight data obtained on day 10 are
  • Tumor values - T/C percent, tumor volume and tumor weight - are significantly lower and body weight gains significantly greater in the rats treated with FT-TAU than in those treated with FT-uracil.
  • FT-TAU hematopoietic effects of FT treatment.
  • Platelet counts are preserved at all FT dose levels in the FT-TAU treated animals, while dropping precipitously at increasing doses of FT in the FT-uracil groups.
  • Total white blood cell counts and lymphocytes are also maintained better in the FT-TAU groups at the higher, more effective doses of FT.
  • neutrophil counts are less severely attenuated in the FT-TAU group than in the equivalent FT-uracil group. In fact, at each FT-TAU dose level, neutrophil counts are approximately twice those observed in the
  • FT-TAU has greater anti-tumor activity than equivalent doses of FT-uracil while causing less intestinal and hematopoietic damage.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Composés, compositions et procédés de traitement et de prévention de la toxicité due à des agents chimiothérapeutiques et à des agents antiviraux; dérivés acylés de nucléosides de pyrimidine non méthylés. Ces composés sont capables d'atténuer les altérations du système hématopoïétique chez des animaux traités par chimiothérapie antivirale ou antinéoplasique.
PCT/US1993/012689 1993-05-14 1993-12-30 Traitement de la toxicite des agents chimioterapeutiques et des agents antiviraux avec des nucleosides de pyrimidine acyles WO1994026761A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU60812/94A AU6081294A (en) 1993-05-14 1993-12-30 Treatment of chemotherapeutic agent and antiviral agent toxicity with acylated pyrimidine nucleosides

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US6138193A 1993-05-14 1993-05-14
US08/061,381 1993-05-14

Publications (1)

Publication Number Publication Date
WO1994026761A1 true WO1994026761A1 (fr) 1994-11-24

Family

ID=22035423

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/012689 WO1994026761A1 (fr) 1993-05-14 1993-12-30 Traitement de la toxicite des agents chimioterapeutiques et des agents antiviraux avec des nucleosides de pyrimidine acyles

Country Status (2)

Country Link
AU (1) AU6081294A (fr)
WO (1) WO1994026761A1 (fr)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5719132A (en) * 1996-06-27 1998-02-17 Bristol-Myers Squibb Company Compositions and methods of treating HIV with d4T, 5-fluorouracil/tegafur, and uracil
EP0831849A1 (fr) * 1995-06-07 1998-04-01 Pro-Neuron, Inc. Procedes de reduction de la toxicite d'agents chimiotherapeutiques et antiviraux avec des nucleosides de pyrimide acyles
FR2763953A1 (fr) * 1997-06-02 1998-12-04 Hoffmann La Roche Derives de 5'-desoxy-cytidine
EP0882734A2 (fr) * 1997-06-02 1998-12-09 F. Hoffmann-La Roche Ag Dérivés de 5'-déoxy-cytidine
WO1999008686A1 (fr) * 1997-08-21 1999-02-25 Basf Aktiengesellschaft Utilisation d'uridine pour contrer la toxicite du 5-fluorouracil
US5945527A (en) * 1996-05-30 1999-08-31 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modification methods using nucleophiles and carbon monoxide
US5959100A (en) * 1996-03-27 1999-09-28 Nexstar Pharmaceuticals, Inc. Pyrimidine nucleosides as therapeutic and diagnostic agents
US6020483A (en) * 1998-09-25 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleoside modifications by palladium catalyzed methods
US6403565B1 (en) 1997-11-04 2002-06-11 Pro-Neuron, Inc. Antimutagenic compositions for treatment and prevention of photodamage to skin
US6784161B2 (en) 2000-02-18 2004-08-31 Biochem Pharma, Inc. Method for the treatment or prevention of flavivirus infections using nucleoside analogues
US6875751B2 (en) 2000-06-15 2005-04-05 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
US7135464B2 (en) 2002-06-05 2006-11-14 Supergen, Inc. Method of administering decitabine
US7250416B2 (en) 2005-03-11 2007-07-31 Supergen, Inc. Azacytosine analogs and derivatives
US7276228B2 (en) 2001-04-24 2007-10-02 Supergen, Inc. Methods for treating hematological disorders through inhibition of DNA methylation and histone deacetylase
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US20100130440A1 (en) * 2001-05-18 2010-05-27 Rakesh Kumar Antiviral nucleosides
EP2265274A2 (fr) * 2008-03-03 2010-12-29 Tosk, Inc. Adjuvants pour réduire la toxicité du méthotrexate, et procédés d utilisation
JP2011500713A (ja) * 2007-10-16 2011-01-06 エイザイ インコーポレイテッド シチジンデアミナーゼ阻害剤としての2’−フルオロ−2’−デオキシテトラヒドロウリジン
EP2295063A2 (fr) 1998-08-31 2011-03-16 32 Mott Street Acquisitions I, LLC d/b/a Wellstat Vaccines Compositions et leur utilisation pour le traitement de maladies mitochondriales
US8324180B2 (en) 2009-04-06 2012-12-04 Eisai Inc. Compositions and methods for treating cancer
US8329665B2 (en) 2009-04-06 2012-12-11 Eisai Inc. Compositions and methods for treating cancer
US8329666B2 (en) 2009-04-06 2012-12-11 Eisai Inc. Compositions and methods for treating cancer
US8609631B2 (en) 2009-04-06 2013-12-17 Eisai Inc. Compositions and methods for treating cancer
US9381207B2 (en) 2011-08-30 2016-07-05 Astex Pharmaceuticals, Inc. Drug formulations
US10485764B2 (en) 2015-07-02 2019-11-26 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US10519190B2 (en) 2017-08-03 2019-12-31 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof
CN112789048A (zh) * 2018-08-14 2021-05-11 托斯克公司 用于治疗粘膜炎的方法和组合物
CN112839663A (zh) * 2018-08-14 2021-05-25 托斯克公司 降低氟尿嘧啶诱发的毒性的方法和组合物
US11446303B2 (en) 2019-06-21 2022-09-20 Tosk, Inc. Uridine phosphorylase (UPase) inhibitors for treatment of liver conditions
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use
US11981703B2 (en) 2016-08-17 2024-05-14 Sirius Therapeutics, Inc. Polynucleotide constructs

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1297398A (fr) * 1969-08-06 1972-11-22
JPS60174797A (ja) * 1984-02-21 1985-09-09 Funai Corp Ν−アロイルチミジン誘導体ならびに抗腫瘍活性物質の毒性低下剤
US4757139A (en) * 1983-07-20 1988-07-12 Teijin Limited 5-fluoro-2'-deoxyuridine derivative, processes for preparing same and antitumor composition containing the same
US4874602A (en) * 1988-02-22 1989-10-17 Paul Calabresi Reduction of the severity 3'-azido-3'-deoxythymidine-induced anemia using benzylacyclouridine
US4950466A (en) * 1988-06-22 1990-08-21 Paul Calabresi Reduction of the severity of 3'-azido-3'-deoxythymidine-induced anemia using a combination of benzylacyclouridine and dipyridamole
US5077290A (en) * 1990-10-11 1991-12-31 Merck & Co., Inc. Morpholine derivatives compositions and use

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1297398A (fr) * 1969-08-06 1972-11-22
US4757139A (en) * 1983-07-20 1988-07-12 Teijin Limited 5-fluoro-2'-deoxyuridine derivative, processes for preparing same and antitumor composition containing the same
JPS60174797A (ja) * 1984-02-21 1985-09-09 Funai Corp Ν−アロイルチミジン誘導体ならびに抗腫瘍活性物質の毒性低下剤
US4874602A (en) * 1988-02-22 1989-10-17 Paul Calabresi Reduction of the severity 3'-azido-3'-deoxythymidine-induced anemia using benzylacyclouridine
US4950466A (en) * 1988-06-22 1990-08-21 Paul Calabresi Reduction of the severity of 3'-azido-3'-deoxythymidine-induced anemia using a combination of benzylacyclouridine and dipyridamole
US5077290A (en) * 1990-10-11 1991-12-31 Merck & Co., Inc. Morpholine derivatives compositions and use

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BIOCHEMICAL PHARMACOLOGY, Vol. 15, issued 1966, CASIDA et al., "3',5'-Diesters of 5-Fluoro-2'-Deoxyuridine and Thymidine Hydrolysis by Esterases in Human, Mouse, and Insect Tissue", pages 627-644. *
BIOCHEMICAL PHARMACOLOGY, Vol. 28, issued 1979, ENSMINGER et al., "Thymidine 5'-O-Pivaloate: A Prodrug Derivative of Thymidine with Potential Applications in High-Dose Methotrexate Therapy", pages 1541-1545. *
CANCER RESEARCH, Vol. 42, issued October 1982, MARTIN et al., "High-Dose 5-Fluorouracil with Delayed Uridine 'Rescue' in Mice", pages 3964-3970. *
CHEMICAL ABSTRACTS, Volume 118, issued 1993, LOSSE et al., "A Convenient Pathway to 2'-(Tert-Butyloxycarbonyl)Ribonucleosides", page 884, Column 1, Abstr. No. 60026c; & J. PRAKT. CHEM./CHEM.-ZTG., 334(6), 531-532. *
JOURNAL OF PHARMACEUTICAL SCIENCES, Vol. 76, No. 2, issued February 1987, MARTIN et al., "Synthesis and Antiviral Activity of Various Esters of 9-Ä(1,3-Dihydroxy-2-Propoxy)MethylÜGuanine", pages 180-183. *

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6344447B2 (en) 1987-10-28 2002-02-05 Pro-Neuron, Inc. Treatment of chemotherapeutic agent and antiviral agent toxicity with acylated pyrimidine nucleosides
EP0831849A1 (fr) * 1995-06-07 1998-04-01 Pro-Neuron, Inc. Procedes de reduction de la toxicite d'agents chimiotherapeutiques et antiviraux avec des nucleosides de pyrimide acyles
JP2009167213A (ja) * 1995-06-07 2009-07-30 Wellstat Therapeutics Corp アシル化ピリミジンヌクレオシドによる化学療法剤および抗ウイルス剤の毒性を減少させる方法
EP1491201A1 (fr) * 1995-06-07 2004-12-29 Wellstat Therapeutics Corporation Procédés de réduction de la toxicité de 5-fluorouracil avec des nucléosides pyrimidiniques acylés
EP0831849A4 (fr) * 1995-06-07 2003-03-26 Wellstat Therapeutics Corp Procedes de reduction de la toxicite d'agents chimiotherapeutiques et antiviraux avec des nucleosides de pyrimide acyles
US5959100A (en) * 1996-03-27 1999-09-28 Nexstar Pharmaceuticals, Inc. Pyrimidine nucleosides as therapeutic and diagnostic agents
US6143882A (en) * 1996-03-27 2000-11-07 Nexstar Pharmaceuticals, Inc. Urea nucleosides as therapeutic and diagnostic agents
US6914138B2 (en) 1996-03-27 2005-07-05 Gilead Sciences, Inc. Urea nucleosides as therapeutic and diagnostic agents
US6441161B1 (en) 1996-03-27 2002-08-27 Gilead Sciences, Inc. Urea nucleosides as therapeutic and diagnostic agents
US5945527A (en) * 1996-05-30 1999-08-31 Nexstar Pharmaceuticals, Inc. Palladium catalyzed nucleoside modification methods using nucleophiles and carbon monoxide
US5719132A (en) * 1996-06-27 1998-02-17 Bristol-Myers Squibb Company Compositions and methods of treating HIV with d4T, 5-fluorouracil/tegafur, and uracil
FR2763953A1 (fr) * 1997-06-02 1998-12-04 Hoffmann La Roche Derives de 5'-desoxy-cytidine
ES2142763A1 (es) * 1997-06-02 2000-04-16 Hoffmann La Roche Derivados de 5'-desoxicitidina.
US6114520A (en) * 1997-06-02 2000-09-05 Hoffmann-La Roche Inc. 5'-deoxy-cytidine derivatives
US6211166B1 (en) * 1997-06-02 2001-04-03 Hoffman-La Roche Inc. 5′-deoxy-cytidine derivative administration to treat solid tumors
EP0882734A2 (fr) * 1997-06-02 1998-12-09 F. Hoffmann-La Roche Ag Dérivés de 5'-déoxy-cytidine
EP0882734A3 (fr) * 1997-06-02 2004-08-25 F. Hoffmann-La Roche Ag Dérivés de 5'-déoxy-cytidine
WO1999008686A1 (fr) * 1997-08-21 1999-02-25 Basf Aktiengesellschaft Utilisation d'uridine pour contrer la toxicite du 5-fluorouracil
US6403565B1 (en) 1997-11-04 2002-06-11 Pro-Neuron, Inc. Antimutagenic compositions for treatment and prevention of photodamage to skin
US6465440B2 (en) 1997-11-04 2002-10-15 Wellstat Therapeutics Corporation Antimutagenic compositions for treatment and prevention of photodamage to skin
US6417170B2 (en) 1997-11-04 2002-07-09 Pro-Neuron, Inc. Antimutagenic compositions for treatment and prevention of photodamage to skin
USRE39573E1 (en) * 1997-11-04 2007-04-17 Wellstat Therapeutics Corporation Antimutagenic compositions for treatment and prevention of photodamage to skin
EP2295063A3 (fr) * 1998-08-31 2011-09-14 Wellstat Therapeutics Corporation Compositions et leur utilisation pour le traitement de maladies mitochondriales
US8067392B2 (en) 1998-08-31 2011-11-29 Wellstat Therapeutics Corporation Compositions and methods for treatment of mitochondrial diseases
EP2295063A2 (fr) 1998-08-31 2011-03-16 32 Mott Street Acquisitions I, LLC d/b/a Wellstat Vaccines Compositions et leur utilisation pour le traitement de maladies mitochondriales
US6512106B2 (en) 1998-09-25 2003-01-28 Gilead Sciences, Inc. Nucleoside modifications by palladium catalyzed methods
US6846918B2 (en) 1998-09-25 2005-01-25 Gilead Sciences, Inc. Nucleoside modifications by palladium catalyzed methods
US6355787B1 (en) 1998-09-25 2002-03-12 Gilead Sciences, Inc. Purine nucleoside modifications by palladium catalyzed methods and compounds produced
US6020483A (en) * 1998-09-25 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleoside modifications by palladium catalyzed methods
US6784161B2 (en) 2000-02-18 2004-08-31 Biochem Pharma, Inc. Method for the treatment or prevention of flavivirus infections using nucleoside analogues
US6875751B2 (en) 2000-06-15 2005-04-05 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
US7585851B2 (en) 2000-06-15 2009-09-08 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
US7276228B2 (en) 2001-04-24 2007-10-02 Supergen, Inc. Methods for treating hematological disorders through inhibition of DNA methylation and histone deacetylase
US20100130440A1 (en) * 2001-05-18 2010-05-27 Rakesh Kumar Antiviral nucleosides
US8227594B2 (en) * 2001-05-18 2012-07-24 Rakesh Kumar Antiviral nucleosides
US7135464B2 (en) 2002-06-05 2006-11-14 Supergen, Inc. Method of administering decitabine
US7144873B2 (en) 2002-06-05 2006-12-05 Supergen, Inc. Kit for delivering decitabine in vivo
US7250416B2 (en) 2005-03-11 2007-07-31 Supergen, Inc. Azacytosine analogs and derivatives
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US10933079B2 (en) 2005-09-29 2021-03-02 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US10456415B2 (en) 2005-09-29 2019-10-29 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US9480698B2 (en) 2005-09-29 2016-11-01 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US8461123B2 (en) 2005-09-29 2013-06-11 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US9358248B2 (en) 2005-09-29 2016-06-07 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US8951987B2 (en) 2007-10-16 2015-02-10 Otsuka Pharmaceuticals Co., Ltd. Certain compounds, compositions and methods
US8268800B2 (en) 2007-10-16 2012-09-18 Eisai Inc. Certain compounds, compositions and methods
US9567363B2 (en) 2007-10-16 2017-02-14 Otsuka Pharmaceutical Co., Ltd. Certain compounds, compositions and methods
JP2011500713A (ja) * 2007-10-16 2011-01-06 エイザイ インコーポレイテッド シチジンデアミナーゼ阻害剤としての2’−フルオロ−2’−デオキシテトラヒドロウリジン
US8618075B2 (en) 2007-10-16 2013-12-31 Eisai Inc. Certain compounds, compositions and methods
US9382287B2 (en) 2008-03-03 2016-07-05 Tosk, Inc. Methotrexate adjuvants to reduce toxicity and methods for using the same
USRE48253E1 (en) 2008-03-03 2020-10-13 Tosk, Inc. Methotrexate adjuvants to reduce toxicity and methods for using the same
EP2265274A4 (fr) * 2008-03-03 2012-01-11 Tosk Inc Adjuvants pour réduire la toxicité du méthotrexate, et procédés d utilisation
US8853227B2 (en) 2008-03-03 2014-10-07 Tosk, Incorporated Methotrexate adjuvants to reduce toxicity and methods for using the same
EP2265274A2 (fr) * 2008-03-03 2010-12-29 Tosk, Inc. Adjuvants pour réduire la toxicité du méthotrexate, et procédés d utilisation
US8609631B2 (en) 2009-04-06 2013-12-17 Eisai Inc. Compositions and methods for treating cancer
US8329665B2 (en) 2009-04-06 2012-12-11 Eisai Inc. Compositions and methods for treating cancer
US8324180B2 (en) 2009-04-06 2012-12-04 Eisai Inc. Compositions and methods for treating cancer
US9040501B2 (en) 2009-04-06 2015-05-26 Otsuka Pharmaceutical Co., Ltd. Compositions and methods for treating cancer
US8329666B2 (en) 2009-04-06 2012-12-11 Eisai Inc. Compositions and methods for treating cancer
US9913856B2 (en) 2011-08-30 2018-03-13 Astex Pharmaceuticals, Inc. Drug formulations
US10517886B2 (en) 2011-08-30 2019-12-31 Astex Pharmaceuticals, Inc. Drug formulations
US9381207B2 (en) 2011-08-30 2016-07-05 Astex Pharmaceuticals, Inc. Drug formulations
US10485764B2 (en) 2015-07-02 2019-11-26 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US11981703B2 (en) 2016-08-17 2024-05-14 Sirius Therapeutics, Inc. Polynucleotide constructs
US11597744B2 (en) 2017-06-30 2023-03-07 Sirius Therapeutics, Inc. Chiral phosphoramidite auxiliaries and methods of their use
US10519190B2 (en) 2017-08-03 2019-12-31 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof
CN112789048A (zh) * 2018-08-14 2021-05-11 托斯克公司 用于治疗粘膜炎的方法和组合物
CN112839663A (zh) * 2018-08-14 2021-05-25 托斯克公司 降低氟尿嘧啶诱发的毒性的方法和组合物
US11446303B2 (en) 2019-06-21 2022-09-20 Tosk, Inc. Uridine phosphorylase (UPase) inhibitors for treatment of liver conditions

Also Published As

Publication number Publication date
AU6081294A (en) 1994-12-12

Similar Documents

Publication Publication Date Title
US5736531A (en) Compositions of chemotherapeutic agent or antiviral agent with acylated pyrimidine nucleosides
US5968914A (en) Treatment of chemotherapeutic agent and antiviral agent toxicity with acylated pyrimidine nucleosides
WO1994026761A1 (fr) Traitement de la toxicite des agents chimioterapeutiques et des agents antiviraux avec des nucleosides de pyrimidine acyles
EP0594667B1 (fr) Traitement d'un agent chimiotherapeutique et de la toxicite d'un agent antiviral par des nucleosides de pyrimidine acylee
US5691320A (en) Acylated pyrimidine nucleosides for treatment of systemic inflammation and inflammatory hepatitis
AU712679B2 (en) Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis
US7776838B1 (en) Treatment of chemotherapeutic agent and antiviral agent toxicity with acylated pyrimidine nucleosides
EP0679160B1 (fr) Precurseurs de nucleotides de pyrimidine utilises pour le traitement de l'hepatite inflammatoire
US6329350B1 (en) Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis
AU2005232281A1 (en) Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis
AU5262499A (en) Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis
AU2491301A (en) Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis
AU2006236108A1 (en) Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP KR

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

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

Ref country code: CA