EP1715871A1 - Kinase inhibitor phosphonate conjugates - Google Patents

Kinase inhibitor phosphonate conjugates

Info

Publication number
EP1715871A1
EP1715871A1 EP04815625A EP04815625A EP1715871A1 EP 1715871 A1 EP1715871 A1 EP 1715871A1 EP 04815625 A EP04815625 A EP 04815625A EP 04815625 A EP04815625 A EP 04815625A EP 1715871 A1 EP1715871 A1 EP 1715871A1
Authority
EP
European Patent Office
Prior art keywords
compound
ofthe
formula
kinase
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP04815625A
Other languages
German (de)
French (fr)
Inventor
Will Watkins
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences 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
Priority claimed from US10/832,811 external-priority patent/US7417055B2/en
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Publication of EP1715871A1 publication Critical patent/EP1715871A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/548Phosphates or phosphonates, e.g. bone-seeking
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention relates generally to phosphonate-containing compounds with kinase-inhibitory activity, i.e., compounds that inhibit at least one kinase.
  • kinase-inhibitory activity i.e., compounds that inhibit at least one kinase.
  • agents currently administered to a patient parenterally are not targeted, thereby resulting in systemic delivery ofthe agent to cells and tissues o he body where the agent is unnecessary, and often undesirable. This systemic delivery may result in adverse side effects and often limits the dose of an agent (e.g., glucocorticoids and other anti-inflammatory agents) that can be administered.
  • an agent e.g., glucocorticoids and other anti-inflammatory agents
  • oral administration of agents is generally recognized as a convenient and economical method of administration.
  • oral administration of agents can result in (a) the uptake ofthe agent through cellular and tissue barriers, such as the blood-brain barrier, epithelial, or the cell membrane, resulting in undesirable systemic distribution, and/or (b) temporary residence ofthe agent within the gastrointestinal tract.
  • a major goal has been to develop methods for specifically targeting agents to cells and tissues.
  • Benefits of such treatment includes avoiding the general physiological effects of inappropriate delivery of such agents to other cells and tissues, such as uninfected cells.
  • therapeutic agents for example, agents that inhibit at least one kinase, with improved pharmacological properties, e.g., drugs having improved kinase-inhibitory activity and pharmacokinetic properties, including improved oral bioavailability, greater potency and extended effective half-life in vivo.
  • Such inhibitors would have therapeutic uses, for example, as anti-cancer agents.
  • new kinase inhibitors should have fewer side effects, less complicated dosing schedules, and be orally active.
  • a less onerous dosage regimen such as one pill, once per day.
  • Assay methods capable of determining the presence, absence or amounts of kinase inhibition are of practical utility in the search for kinase inhibitors as well as for diagnosing the presence of conditions associated with kinase activity.
  • Intracellular targeting may be achieved by methods and compositions that allow accumulation or retention of biologically active agents inside cells.
  • the present invention provides novel analogs of kinase-inhibitory compounds, i.e., compounds that inhibit the activity of at least one kinase.
  • Such novel kinase-inhibitory analogs possess utilities ofthe kinase-inhibitory compounds and optionally provide cellular accumulation.
  • the present invention provides compositions and methods useful for inhibiting at least one kinase that may have therapeutic activity against diseases associated with kinase activity, such as cancer.
  • the present invention relates generally to the accumulation or retention of therapeutic compounds inside cells.
  • the invention is more particularly related to attaining high concentrations of phosphonate-containing molecules in target cells. Such effective targeting may be applicable to a variety of therapeutic formulations and procedures.
  • Compounds ofthe invention include kinase-inhibitory compounds having at least one phosphonate group. Accordingly, in one embodiment the invention provides a conjugate comprising a kinase inhibiting compound linked to one or more phosphonate groups; or a pharmaceutically acceptable salt or solvate thereof. In another embodiment the invention provides a compound comprising one or more phosphonates and a substructure of formula I:
  • L 1 and L 2 are -N- or -CR a -; and R a is hydrogen, alkyl, substituted alkyl, aryl or substituted aryl; or a pharmaceutically acceptable salt thereo.
  • the invention provides compound comprising one or more phosphonates and a substructure of formula II:
  • the invention provides compound comprising one or more phosphonates and a substructure of formula Ilia, INa or Va: ma IVa Va
  • the invention provides compound comprising one or more phosphonates and a substructure of formula III, IN or N:
  • the invention provides a compound of any one of formulae 1-4:
  • a A 0 is A 1 ;
  • a 1 is
  • a 3 is:
  • Y 1 is independently O, S, N(R X ), N(OR x ), or N(N(R X )( R x ));
  • Y 2 is independently a bond, O, N(R X ), N(OR x ), N(N(R X )( R x )), or - S(O) M 2 ⁇ ; and when Y 2 joins two phosphorous atoms Y 2 can also be C(R 2 )(R 2 );
  • R x is independently H, R , W , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group;
  • R 2 is independently H, R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R 3 groups;
  • R 3 is R 3a , R 3b , R 3 ° or R 3d , provided that when R 3 is bound to a heteroatom, then R 3 is R 3c or R 3d ;
  • R 3a is F, Cl, Br, I, -CN, N 3 or -NO 2 ;
  • R 3b is Y 1 ;
  • R 3c is -R x , -N(R X )(R X ), -SR X , -S(O)R x , -S(O) 2 R x , -S(O)(OR x ), - S(O) 2 (OR x ), -OC(Y 1 )R x , -OC(Y 1 )OR x , -OC(Y 1
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound ofthe invention and a pharmaceutically acceptable excipient.
  • the invention also provides a method of increasing cellular accumulation and retention of drug compounds, thus improving their therapeutic and diagnostic value, comprising linking the compound to one or more (e.g., 1, 2, 3, or 4) phosphonate groups.
  • the invention provides a method of inhibiting the activity of at least one kinase in an animal (e.g. a mammal) comprising administering an effective amount of a compound ofthe invention to the animal.
  • the invention provides a unit dosage form comprising a compound ofthe invention and a pharmaceutically acceptable excipient.
  • the invention provides a method for inhibiting a kinase in vitro or in vivo comprising contacting a sample in need of such treatment with a compound ofthe invention.
  • the invention provides a method of treating cancer in an animal (e.g. a mammal) in need of such treatment comprising administering an effective amount of a compound ofthe invention to the animal.
  • the invention provides a compound ofthe invention for use in medical therapy (preferably for use in treating a condition associated with kinase activity, e.g., elevated kinase activity), as well as the use of a compound of the invention for the manufacture of a medicament useful for the treatment of a condition associated with kinase activity, e.g., associated with elevated kinase activity.
  • the invention provides the use of a compound as described in any one of claims 1-55 to prepare a medicament for inhibiting a kinase in an animal (e.g. a mammal).
  • the invention provides the use of a compound of the invention to prepare a medicament for treating cancer in an animal (e.g. a mammal).
  • the invention provides a method for preparing a compound ofthe invention as described in the schemes and examples herein.
  • the invention provides a method for preparing a pharmaceutical composition, comprising combining a pharmaceutically acceptable excipient and a compound ofthe invention.
  • the invention provides processes and novel intermediates disclosed herein which are useful for preparing compounds of he invention. Some ofthe compounds ofthe invention are useful to prepare other compounds ofthe invention.
  • the activity of a kinase is inhibited by a method comprising the step of treating a sample suspected of containing a kinase with a compound or composition of the invention.
  • Receptor tyrosine kinases are important in the transmission of biochemical signals that initiate cell replication. They are large enzymes that span the cell membrane and possess an extracellular binding domain for growth factors such as epidermal growth factor (EGF), and an intracellular portion that functions as a kinase to phosphorylate tyrosine amino acids in proteins and hence influence cell proliferation.
  • EGF epidermal growth factor
  • Class I receptor tyrosine kinases comprising the EGF family of receptor tyrosine kinases such as the EGF, TGF ⁇ , NEU, erbB, Xmrk, HER and let23 receptors
  • Class II receptor tyrosine kinases comprising the insulin family of receptor tyrosine kinases such as the insulin, IGFI and insulin- related receptor (IRR) receptors
  • Class III receptor tyrosine kinases comprising the platelet-derived growth factor (PDGF) family of receptor tyrosine kinases such as the PDGF ⁇ , PDGF ⁇ and colony-stimulating factor I (CSF1) receptors.
  • PDGF platelet-derived growth factor
  • Class I kinases such as the EGF family of receptor tyrosine kinases, are frequently present in common human cancers such as breast cancer (Sainsbury et al, Brit. J. Cancer, 1988, 58, 458; Guerin et al, Oncogene Res., 1988, 3, 21 and Klijn et al, Breast Cancer Res. Treat, 1994, 29, 73), non-small cell lung cancers (NSCLCs) including adenocarcinomas (Cerny et al, Brit. J. Cancer, 1986, 54, 265; Reubi et al, Int. J. Cancer, 1990, 45, 269; and Rusch et al, Cancer Research, 1993, 53, 2379) and squamous cell cancer ofthe lung
  • EGF type tyrosine kinase activity is rarely detected in normal cells, whereas it is more frequently detected in malignant cells (Hunter, Cell, 1987, 50, 823).
  • EGF receptors that possess tyrosine kinase activity are overexpressed in many human cancers such as brain, lung squamous cell, bladder, gastric, breast, head and neck, oesophageal, gynaecological and thyroid tumors (W. J. Gullick, Brit. Med. Bull, 1991, 47, 87). Accordingly, an inhibitor of receptor tyrosine kinases would be of value as a selective inhibitor ofthe growth of mammalian cancer cells (Yaish et al.
  • kinase inhibitors have valuable pharmacological properties and can be used, for example, as anti-tumor drugs and as drugs against atherosclerosis.
  • the phosphorylation of proteins has long been known as an important step in the differentiation and proliferation of cells. Phosphorylation is catalyzed by protein kinases that are divided into serine/threonine kinases and tyrosine kinases.
  • the serine/threonine kinases include protein kinase C and the tyrosine kinases include PDGF (platelet-derived growth factor)-receptor tyrosine kinase and Bcr- Abl kinase.
  • Chronic myelogenous Leukemia is a hematological stem cell disorder associated with a specific chromosomal translocation known as the Philadelphia chromosome that is detected in 95% of patients with CML and 20% with acute lymphocytic leukemia (ALL).
  • ALL acute lymphocytic leukemia
  • the molecular consequences ofthe translocation is the fusion ofthe abl protooncogene to the bcr gene resulting in the production of an activated from of Abl tyrosine protein kinase.
  • the Bcr-Abl protein is capable of inducing leukemias in mice, thus implicating the protein as the cause of these diseases.
  • kinase inhibitors inhibit cellular kinases that are involved in disease states, for example, Bcr-Abl.
  • kinase inhibitors prevent the development of resistance (multi-drug resistance) in cancer treatment with other chemotherapeutic drugs or remove existing resistance to other chemotherapeutic drugs.
  • Two processes, the de novo formation of vessels from differentiating endothelial cells or angioblasts in the developing embryo (vasculogenesis) and the growth of new capillary vessels from existing blood vessels (angiogenesis) are involved in the development ofthe vascular systems of animal organs and tissues.
  • Transient phases of new vessel formation also occur in the adult body, for example, during the menstrual cycle, pregnancy and wound healing.
  • angiogenesis a number of diseases are known to be associated with deregulated angiogenesis, for example, retinopathies, psoriasis, hemangioblastoma, hemangioma, and neoplastic diseases (e.g., solid tumors).
  • retinopathies retinopathies, psoriasis, hemangioblastoma, hemangioma, and neoplastic diseases (e.g., solid tumors).
  • vasculogenesis and angiogenesis have been found to involve a whole range of molecules, especially angiogenic growth factors and their endothelial receptors, as well as cell adhesion molecules.
  • VEGF vascular endothelial growth factor
  • PDGF platelet-derived growth factor
  • VEGF vascular endothelial growth factor
  • monocytes chemotactic for endothelial cells and monocytes, and induces plasminogen activators in endothelial cells, which are then involved in the proteolytic degradation of extracellular matrix during the formation of capillaries.
  • a number of isoforms of VEGF show comparable biological activity, but differ in the type of cells that secrete them and in their heparin-binding capacity.
  • PLGF placenta growth factor
  • VEGF receptors are transmembranous receptor tyrosine kinases. They are characterized by an extracellular domain with seven immunoglobulin-like domains and an intracellular tyrosine kinase domain.
  • VEGF receptor Various types of VEGF receptor are known, e.g. VEGFR-1, VEGFR-2, and VEGFR-3.
  • Direct evidence ofthe role of VEGF as a tumor angiogenesis factor in vivo has been obtained from studies in which VEGF expression or VEGF activity was inhibited.
  • Epidermal Growth Factor, Transforming Growth Factor a, Transforming Growth Factor A, Interleukin 1, and Interleukin 6, induce the expression of VEGF in cell experiments.
  • Angiogenesis is regarded as a prerequisite for those tumors that grow beyond a maximum diameter of about 1-2 mm; up to this limit, oxygen and nutrients may be supplied to the tumor cells by diffusion. Every tumor, regardless of its origin and its cause, is thus thought to be dependent on angiogenesis for its growth after it has reached a certain size.
  • kinase inhibitors with improved pharmacological properties, e.g. , drugs having improved kinase- inhibitory activity and pharmacokinetic properties, including improved oral bioavailability, greater potency and extended effective half-life in vivo.
  • Such inhibitors would have therapeutic potential as, e.g., anticancer agents.
  • the kinase inhibitory compounds provided herein may be used to treat breast cancer, non-small cell lung cancers (NSCLCs), adenocarcinomas, squamous cell cancer ofthe lung, oesophageal cancer, gastrointestinal cancer, colon cancer, rectal cancer, stomach cancer, prostate cancer, leukaemia, ovarian cancer, bronchial cancer, pancreatic cancer, thyroid cancer, uterine cancer, brain cancer, lung squamous cell cancer, bladder cancer, gastric cancer, head and neck cancer, gynaecological and thyroid tumors, to prevent the development of resistance (multi-drug resistance) in cancer treatment with other chemotherapeutic drugs or remove existing resistance to other chemotherapeutic drugs, retinopathies, hemangioblastoma, hemangioma, and neoplastic diseases, gliomas, to inhibit tumor angiogenesis, myelomas, chronic myeloid leukemia (CML), acute lymphocytic leukemia
  • phosphonate and “phosphonate group” include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to a heteroatom , 3) single-bonded to a heteroatom, and 4) single-bonded to another heteroatom, wherein each heteroatom can be the same or different.
  • phosphonate and “phosphonate group” also include functional groups or moieties that comprise a phosphorous in the same oxidation state as the phosphorous described above, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having the characteristics described above.
  • the terms “phosphonate” and “phosphonate group” include phosphonic acid, phosphonic monoester, phosphonic diester, phosphonamidate, and phosphonthioate functional groups.
  • the terms “phosphonate” and “phosphonate group” include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double- bonded to an oxygen, 3) single-bonded to an oxygen, and 4) single-bonded to another oxygen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteristics.
  • the terms "phosphonate” and “phosphonate group” include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to an oxygen, 3) single-bonded to an oxygen or nitrogen, and 4) single-bonded to another oxygen or nitrogen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteristics.
  • prodrug refers to any compound that when administered to a biological system generates the drug substance, i.e.
  • prodrug is thus a covalently modified analog or latent form of a therapeutically- active compound.
  • Prodrug moiety refers to a labile functional group that separates from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, “Design and Application of Prodrugs” in A Textbook of Drug Design and Development (1991), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academic Publishers, pp. 113-191).
  • Enzymes that are capable of an enzymatic activation mechanism with the phosphonate prodrug compounds ofthe invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphases.
  • Prodrug moieties can serve to enhance solubility, absorption and lipophilicity to optimize drug delivery, bioavailability and efficacy.
  • a prodrug moiety may include an active metabolite or drug itself.
  • the acyloxyalkyl ester was first used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al. (1983) J. Pharm. Sci. 72: 324; also U.S. Patent Nos.
  • acyloxyalkyl ester was used to deliver phosphonic acids across cell membranes and to enhance oral bioavailability.
  • a close variant ofthe acyloxyalkyl ester, the alkoxycarbonyloxyalkyl ester (carbonate), may also enhance oral bioavailability as a prodrug moiety in the compounds ofthe combinations ofthe invention.
  • the phosphonate group may be a phosphonate prodrug moiety.
  • the prodrug moiety may be sensitive to hydrolysis, such as, but not limited to a pivaloyloxymethyl carbonate (POC) or POM group.
  • the prodrug moiety may be sensitive to enzymatic potentiated cleavage, such as a lactate ester or a phosphonamidate-ester group.
  • Aryl esters of phosphorus groups are reported to enhance oral bioavailability (De Lombaert et al. (1994) J. Med. Chem. 37: 498). Phenyl esters containing a carboxylic ester ortho to the phosphate have also been described (Khamnei and Torrence, (1996) J. Med. Chem. 39:4109- 4115). Benzyl esters are reported to generate the parent phosphonic acid. In some cases, substituents at the ort/ * o-or/> ⁇ r ⁇ -position may accelerate the hydrolysis.
  • Benzyl analogs with an acylated phenol or an alkylated phenol may generate the phenolic compound through the action of enzymes, e.g., esterases, oxidases, etc., which in turn undergoes cleavage at the benzylic C-O bond to generate the phosphoric acid and the quinone methide intermediate.
  • enzymes e.g., esterases, oxidases, etc.
  • this class of prodrugs are described by Mitchell et al. (1992) J. Chem. Soc. Perkin Trans. II 2345; Glazier WO 91/19721.
  • Still other benzylic prodrugs have been described containing a carboxylic ester-containing group attached to the benzylic methylene (Glazier WO 91/19721).
  • Thio-containing prodrugs are reported to be useful for the intracellular delivery of phosphonate drugs. These proesters contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide.
  • Protecting Groups in Organic Chemistry Theodora W. Greene, John Wiley & Sons, Inc., New York, 1991.
  • Protecting groups are often utilized to mask the reactivity of certain functional groups, to assist in the efficiency of desired chemical reactions, e.g. , making and breaking chemical bonds in an ordered and planned fashion. Protection of functional groups of a compound alters other physical properties besides the reactivity of the protected functional group, such as the polarity, lipophilicity (hydrophobicity), and other properties which can be measured by common analytical tools.
  • Chemically protected intermediates may themselves be biologically active or inactive.
  • Protected compounds may also exhibit altered, and in some cases, optimized properties in vitro and in vivo, such as passage through cellular membranes and resistance to enzymatic degradation or sequestration. In this role, protected compounds with intended therapeutic effects may be referred to as prodrugs.
  • Another function of a protecting group is to convert the parental drug into a prodrug, whereby the parental drug is released upon conversion of the prodrug in vivo. Because active prodrugs may be absorbed more effectively than the parental drug, prodrugs may possess greater potency in vivo than the parental drug.
  • Protecting groups are removed either in vitro, in the instance of chemical intermediates, or in vivo, in the case of prodrugs.
  • any reference to any ofthe compounds ofthe invention also includes a reference to a physiologically acceptable salt thereof.
  • physiologically acceptable salts ofthe compounds ofthe invention include salts derived from an appropriate base, such as an alkali metal (for example, sodium), an alkaline earth (for example, magnesium), ammonium and NX 4 + (wherein X is C1-C4 alkyl).
  • Physiologically acceptable salts of a hydrogen atom or an amino group include salts of organic carboxylic acids such as acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, sulfuric, phosphoric and sulfamic acids.
  • organic carboxylic acids such as acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids
  • organic sulfonic acids such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids
  • Physiologically acceptable salts of a compound of an hydroxy group include the anion of said compound in combination with a suitable cation such as Na + and NX (wherein X is independently selected from H or a C 1 -C 4 alkyl group).
  • a suitable cation such as Na + and NX (wherein X is independently selected from H or a C 1 -C 4 alkyl group).
  • salts of active ingredients ofthe compounds ofthe invention will be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base.
  • salts of acids or bases which are not physiologically acceptable may also find use, for example, in the preparation or purification of a physiologically acceptable compound. All salts, whether or not derived form a physiologically acceptable acid or base, are within the scope ofthe present invention.
  • the term "substructure” refers to a residue wherein any hydrogen atom(s) or replaceable group(s) has been or can be removed to provide an open valence for the substitution of a group including a phosphonate group, e.g., the substructure is a scaffold, to which a substituent -link-P(O)(OR 1 ) 2 is attached.
  • the substructures can have additional groups attached.
  • a kinase inhibiting compound that comprises at least one phosphonate group and a substructure it is understood that the compound includes the substructure as at least part ofthe overall structure ofthe compound.
  • Alkyl is C1-C18 hydrocarbon containing normal, secondary, or tertiary carbon atoms.
  • Examples are methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1- propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-pro ⁇ yl (i-Pr, i-propyl, -CH(CH3)2), 1 -butyl (n-Bu, n-butyl, -CH2CH2CH2CH3), 2-methyl-l -propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, -CH(CH3)CH2CH3), 2-methyl-2- propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, -CH2CH2CH2CH3, 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-
  • Alkynyl is C2-C18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. Examples include, but are not limited to, acetylenic (-C ⁇ CH) and propargyl (-CH 2 C ⁇ CH).
  • Alkylene refers to a saturated, branched or straight chain or cyclic hydrocarbon radical of 1-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane.
  • alkylene radicals include, but are not limited to, methylene (-CH 2 -) 1,2-ethyl (-CH 2 CH 2 -), 1,3-propyl (-CH 2 CH 2 CH 2 -), 1,4-butyl (-CH 2 CH 2 CH 2 CH 2 -), and the like.
  • Alkenylene refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene.
  • Alkynylene refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne.
  • Typical alkynylene radicals include, but are not limited to, acetylene (-C ⁇ C-), propargyl (-CH 2 C ⁇ C-), and 4-pentynyl (-CH 2 CH 2 CH 2 C ⁇ CH-).
  • Aryl means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
  • Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like.
  • “Arylalkyl” refers to an acyclic alkyl radical in which one ofthe hydrogen atoms bonded to a carbon atom, typically a terminal or sp carbon atom, is replaced with an aryl radical.
  • Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-l-yl, , naphthylmethyl, 2-naphthylethan-l- yl, naphthobenzyl, 2-naphthophenylethan-l-yl and the like.
  • the arylalkyl group comprises 6 to 20 carbon atoms, e.g., the alkyl moiety, including alkanyl, alkenyl or alkynyl groups, ofthe arylalkyl group is 1 to 6 carbon atoms and the aryl moiety is 5 to 14 carbon atoms.
  • substituted cycloalkyl mean alkyl, aryl, arylalkyl, and cycloalkyl respectively, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent.
  • Heterocycle as used herein includes, by way of example and not limitation, those heterocycles described in Paquette, Leo A.; Principles of Modern Heterocyclic Chemistrv (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; The Chemistrv of Heterocyclic Compounds. A Series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; andJ. Am. Chem. Soc. (1960) 82:5566.
  • heterocycle includes a “carbocycle” as defined herein, wherein one or more (e.g., 1, 2, 3, or 4) carbon atoms have been replaced with a heteroatom (e.g., O, N, or S).
  • a heteroatom e.g., O, N, or S.
  • heterocycles include, by way of example and not limitation, pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolin
  • carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
  • carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5- pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2- pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3- pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
  • nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1 -pyrrolyl, 1 -imidazolyl, 1- pyrazolyl, and 1-piperidinyl.
  • Carbocycle refers to a saturated, unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle.
  • Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms.
  • Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyclo [5,6] or [6,6] system.
  • Examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1- cyclopent-1-enyl, l-cyclo ⁇ ent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1- cyclohex-1-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, phenyl, spiryl and naphfhyl.
  • cycloalkyl refers to a Ci-Cis hydrocarbon containing one or more rings.
  • chiral refers to molecules which have the property of non- superimposability ofthe mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement ofthe atoms or groups in space.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non- superimposable mirror images of one another.
  • treatment or “treating,” to the extent it relates to a disease or condition includes preventing the disease or condition from occurring, inhibiting the disease or condition, eliminating the disease or condition, and/or relieving one or more symptoms ofthe disease or condition.
  • Stereochemical definitions and conventions used herein generally follow S. P. Parker. Ed.. McGraw-Hill Dictionary of Chemical Terms (19841 McGraw- Hill Book Company, New York; and Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • the terms "racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • protecting groups include prodrug moieties and chemical protecting groups. Protecting groups are available, commonly known and used, and are optionally used to prevent side reactions with the protected group during synthetic procedures, i.e.
  • protecting groups for -OH groups include "ether- or ester- forming groups”.
  • Ether- or ester-forming groups are capable of functioning as chemical protecting groups in the synthetic schemes set forth herein.
  • some hydroxyl and thio protecting groups are neither ether- nor ester-forming groups, as will be understood by those skilled in the art, and are included with amides, discussed below.
  • Ester-forming groups include: (1) phosphonate ester-forming groups, such as phosphonamidate esters, phosphorothioate esters, phosphonate esters, and phosphon-bis-amidates; (2) carboxyl ester-forming groups, and (3) sulphur ester-forming groups, such as sulphonate, sulfate, and sulfinate.
  • the phosphonate moieties ofthe compounds ofthe invention may or may not be prodrug moieties, i.e. they may or may be susceptible to hydrolytic or enzymatic cleavage or modification. Certain phosphonate moieties are stable under most or nearly all metabolic conditions.
  • a dialkylphosphonate where the alkyl groups are two or more carbons, may have appreciable stability in vivo due to a slow rate of hydrolysis.
  • phosphonate prodrug moieties a large number of structurally-diverse prodrugs have been described for phosphonic acids (Freeman and Ross in Progress in Medicinal Chemistry 34: 112-147 (1997) and are included within the scope ofthe present invention.
  • An exemplary phosphonate ester-forming group is the phenyl carbocycle in substructure A 3 having the formula:
  • Ri maybe H or C -Cn alkyl; ml is 1, 2, 3, 4, 5, 6, 7 or 8, and the phenyl carbocycle is substituted with 0 to 3 R 2 groups.
  • Yi is O, a lactate ester is formed, and where Yi is N(R ), N(OR 2 ) or N(N(R 2 ) 2 , a phosphonamidate ester results.
  • a protecting group typically is bound to any acidic group such as, by way of example and not limitation, a -CO2H or
  • R x for example includes the enumerated ester groups of WO 95/07920.
  • protecting groups include: C3-C12 heterocycle (described above) or aryl. These aromatic groups optionally are polycyclic or monocyclic.
  • Examples include phenyl, spiryl, 2- and 3-pyrrolyl, 2- and 3-thienyl, 2- and 4-imidazolyl, 2-, 4- and 5-oxazolyl, 3- and 4-isoxazolyl, 2-, 4- and 5-thiazolyl, 3-, 4- and 5-isothiazolyl, 3- and 4- pyrazolyl, 1-, 2-, 3- and 4-pyridinyl, and 1-, 2-, 4- and 5-pyrimidinyl, C3-C12 heterocycle or aryl substituted with halo, R 1 , R ⁇ O-Ci-C ⁇ alkylene, C1-C12 alkoxy, CN, NO2, OH, carboxy, carboxyester, thiol, thioester, C1-C12 haloalkyl (1-6 halogen atoms), C2-C12 alkenyl or C2-C12 alkynyl.
  • Such groups include 2-, 3- and 4-alkoxyphenyl (C1-C12 alkyl), 2-, 3- and 4- methoxyphenyl, 2-, 3- and 4-ethoxyphenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5- diethoxyphenyl, 2- and 3-carboethoxy-4-hydroxyphenyl, 2- and 3-ethoxy-4- hydroxyphenyl, 2- and 3-ethoxy-5-hydroxyphenyl, 2- and 3-ethoxy-6- hydroxyphenyl, 2-, 3- and 4-O-acetylphenyl, 2-, 3- and 4-dimethylaminophenyl, 2-, 3- and 4-methylmercaptophenyl, 2-, 3- and 4-halophenyl (including 2-, 3- and 4-fluorophenyl and 2-, 3- and 4-chlorophenyl), 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5-dimethylphenyl, 2,3-, 2,4-, 2,5-,
  • esters of 2-carboxyphenyl and C 1 -C 4 alkylene-C 3 -C 6 aryl (including benzyl, - CH 2 -pyrrolyl, -CH 2 -thienyl, -CH 2 -imidazolyl, -CH 2 -oxazolyl, -CH 2 -isoxazolyl, -CH 2 -thiazolyl, -CH 2 -isothiazolyl, -CH 2 -pyrazolyl, -CH 2 -pyridinyl and -CH 2 - pyrimidinyl) substituted in the aryl moiety by 3 to 5 halogen atoms or 1 to 2 atoms or groups selected from halogen, C 1 -C 12 alkoxy (including methoxy and ethoxy), cyano, nitro, OH, C 1 -C 12 haloalkyl (1 to 6 halogen atoms; including - CH 2 CCI3), C 1
  • C ⁇ .is or CO- 10 fatty acids such as linoleic, lauric, myristic, palmitic, stearic, oleic, palmitoleic, linolenic and the like fatty acids
  • fatty acids such as linoleic, lauric, myristic, palmitic, stearic, oleic, palmitoleic, linolenic and the like fatty acids
  • acyl ofthe parental compounds herein through a glyceryl oxygen ofthe triglyceride
  • phospholipids linked to the carboxyl group through the phosphate ofthe phospholipid phthalidyl (shown in Fig. 1 of Clayton et al, Antimicrob. Agents Chemo.
  • hydroxyl groups ofthe compounds of this invention optionally are substituted with one of groups III, IV or V disclosed in WO 94/21604, or with isopropyl.
  • Table A lists examples of protecting group ester moieties that for example can be bonded via oxygen to -C(O)O- and -P(O)(O-)2 groups.
  • the esters of structures 5-7, 11, 12, 21, and 23-26 are synthesized by reaction ofthe alcohol or alkoxide salt (or the corresponding amines in the case of compounds such as 13, 14 and 15) with the monochlorophosphonate or dichlorophosphonate (or another activated phosphonate).
  • # - chiral center is (R), (S) or racemate.
  • Other esters that are suitable for use herein are described in EP 632048.
  • Protecting groups also includes "double ester" forming profunctionalities
  • alkyl- or aryl-acyloxyalkyl groups ofthe structure -CH(R 1 or W 5 )O((CO)R 37 ) or -CH(R 1 or W 5 )((CO)OR 38 ) (linked to oxygen ofthe acidic group) wherein R 37 and R 38 are alkyl, aryl, or alkylaryl groups (see U.S. Patent No. 4968788).
  • R 37 and R 38 are bulky groups such as branched alkyl, ortho- substituted aryl, meta-substituted aryl, or combinations thereof, including normal, secondary, iso- and tertiary alkyls of 1-6 carbon atoms.
  • An example is the pivaloyloxymethyl group.
  • Such useful protecting groups are alkylacyloxymethyl esters and their derivatives, including -
  • the protected acidic group is an ester ofthe acidic group and is the residue of a hydroxyl-containing functionality.
  • an amino compound is used to protect the acid ftxnctionality.
  • the residues of suitable hydroxyl or amino-containing functionalities are set forth above or are found in WO 95/07920. Of particular interest are the residues of amino acids, amino acid esters, polypeptides, or aryl alcohols. Typical amino acid, polypeptide and carboxyl-esterified amino acid residues are described on pages 11 -18 and related text of WO 95/07920 as groups LI or L2.
  • esters of unsubstituted aryl such as phenyl or arylalkyl such benzyl, or hydroxy-, halo-, alkoxy-, carboxy- and/or alkylestercarboxy-substituted aryl or alkylaryl, especially phenyl, ortho-ethoxyphenyl, or C1-C4 alkylestercarboxyphenyl (salicylate C1-C12 alkylesters).
  • the protected acidic groups particularly when using the esters or amides of WO 95/07920, are useful as prodrugs for oral administration. However, it is not essential that the acidic group be protected in order for the compounds of this invention to be effectively administered by the oral route.
  • the compounds ofthe invention having protected groups in particular amino acid amidates or substituted and unsubstituted aryl esters are administered systemically or orally they are capable of hydrolytic cleavage in vivo to yield the free acid.
  • One or more ofthe acidic hydroxyls are protected. If more than one acidic hydroxyl is protected then the same or a different protecting group is employed, e.g., the esters may be different or the same, or a mixed amidate and ester may be used.
  • Typical hydroxy protecting groups described in Greene (pages 14-118) include substituted methyl and alkyl ethers, substituted benzyl ethers, silyl ethers, esters including sulfonic acid esters, and carbonates. For example:
  • Methyl Ethers (Methoxymethyl, Methylthiomethyl, t- Butylthiomethyl, (Phenyldimethylsilyl)methoxymethyl, Benzyloxymethyl, p- Methoxybenzyloxymethyl, (4-Methoxyphenoxy)methyl, Guaiacolmethyl, t- Butoxymethyl, 4-Pentenyloxymethyl, Siloxymethyl, 2- Methoxyethoxymethyl, 2,2,2-Trichloroethoxymethyl, Bis(2- chloroethoxy)methyl, 2-(Trimethylsilyl)ethoxymefhyl, Tetrahydropyranyl, 3- Bromotetrahydropyranyl, Tetrahydropthiopyranyl, 1-Methoxycyclohexyl, 4- Methoxytetrahydropyranyl, 4-Methoxytetrahydrothiopyranyl, 4- Methoxytetrahydropthiopyranyl S,S-Dioxido, l-
  • Ethyl Ethers (1-Ethoxyethyl, 1 -(2-Chloroethoxy)ethyl, 1 -Methyl- 1-methoxyethyl, 1 -Methyl- 1-benzyloxyethyl, 1 -Methyl- l-benzyloxy-2- fluoroethyl, 2,2,2-Trichloroethyl, 2-Trimethylsilylethyl, 2- (Phenylselenyl)ethyl,
  • Esters (Formate, Benzoylformate, Acetate, Choroacetate, Dichloroacetate, Trichloroacetate, Trifluoroacetate, Methoxyacetate, Triphenylmethoxyacetate, Phenoxyacetate, --Chlorophenoxyacetate, p-poly- Phenylacetate, 3-Phenylpropionate, 4-Oxopentanoate (Levulinate), 4,4- (Ethylenedithio)pentanoate, Pivaloate, Adamantoate, Crotonate, 4- Methoxycrotonate, Benzoate, -Phenylbenzoate, 2,4,6-Trimethylbenzoate (Mesitoate));
  • Typical 1,2-diol protecting groups are described in Greene at pages 118-142 and include Cyclic Acetals and Ketals (Methylene, Ethylidene, 1- t-Butylethylidene, 1-Phenylethylidene, (4-Methoxyphenyl)ethylidene, 2,2,2- Trichloroethylidene, Acetonide (Isopropylidene), Cyclopentylidene, Cyclohexylidene, Cycloheptylidene, Benzylidene,/ * -Methoxybenzylidene, 2,4- Dimethoxybenzylidene, 3,4-Dimethoxybenzylidene, 2- ⁇ itrobenzylidene); Cyclic Ortho Esters (Methoxymethylene, Ethoxymethylene, Dimethoxymethylene, 1- Methoxyethylidene, 1-Ethoxye
  • 1,2-diol protecting groups include those shown in Table B, still more typically, epoxides, acetonides, cyclic ketals and aryl acetals.
  • R ⁇ is C1-C6 alkyl
  • Amino protecting groups Another set of protecting groups include any ofthe typical amino protecting groups described by Greene at pages 315-385. They include:
  • Ethyl (2,2,2-trichoroethyl, 2-trimethylsilylethyl, 2-phenylethyl, 1 -(1 -adamantyl)- 1 -methylethyl, 1 , 1 -dimethyl-2-haloethyl, 1 , 1 -dimethyl-2,2- dibromoethyl, l,l-dimethyl-2,2,2-trichloroethyl, 1 -methyl- 1 -(4- bi ⁇ henylyl)ethyl, 1 -(3, 5-di-t-butylphenyl)-l -methylethyl, 2-(2'- and 4*- pyridyl)ethyl, 2-(NN-dicyclohexylcarboxamido)ethyl, t-butyl, 1-adamantyl, vinyl, allyl, 1-isopropylallyl, cinnamyl, 4-
  • N- Alkyl and N-Aryl Amines (N-methyl, N-allyl, N-[2- (trimethylsilyl)ethoxy]methyl, N-3-acetoxypro ⁇ yl, N-(l-isopropyl-4-nitro-2- oxo-3 -pyrrolin-3-yl), Quaternary Ammonium Salts, N-benzyl, N-di(4- methoxyphenyl)methyl, N-5-dibenzosuberyl, N-triphenylmethyl, N-(4- methoxyphenyl)diphenylmethyl, N-9-phenylfluorenyl, N-2,7-dichloro-9- fluorenylmethylene, N-ferrocenylmethyl, N-2-picolylamine N-oxide);
  • N-benzenesulfenyl N-o-nitrobenzenesulfenyl, N-2,4-dinitrobenzenesulfenyl, N- pentachlorobenzenesulfenyl, N-2-nitro-4-methoxybenzenesulfenyl, N- triphenylmethylsulfenyl, N-3-nitropyridinesulfenyl
  • N-sulfonyl Derivatives N-p-toluenesulfonyl, N-benzenesulfonyl, N-2,3,6-trimethyl-4- methoxybenzenesulfonyl, N-2,4,6-trimethoxybenzenesulfonyl, N-2,6- dimethyl-4-methoxybenzenesulfonyl, N-p
  • Another protecting group, also useful as a prodrug for amino or -NH(R 5 ), is:
  • Amino acid and polypeptide protecting group and conjugates An amino acid or polypeptide protecting group of a compound ofthe invention has the structure R 15 NHCH(R 16 )C(O)-, where R 15 is H, an amino acid or polypeptide residue, or R 5 , and R 16 is defined below.
  • R 16 is lower alkyl or lower alkyl (Ci-C ⁇ ) substituted with amino, carboxyl, amide, carboxyl ester, hydroxyl, C 6 -C 7 aryl, guanidinyl, imidazolyl, indolyl, sulfhydryl, sulfoxide, and/or alkylphosphate.
  • R 10 is generally the side group of a naturally-occurring amino acid such as H, - CH 3 , -CH(CH 3 ) 2 , -CH 2 -CH(CH 3 ) 25 -CHCH3-CH2-CH3, -CH 2 -C 6 H 5 , -CH 2 CH 2 - S-CH3, -CH 2 OH, -CH(OH)-CH 3 , -CH 2 -SH, -CH 2 -C 6 H 4 OH, -CH 2 -CO-NH 2 , - CH2-CH2-CO-NH2, -CH2-COOH, -CH2-CH2-COOH, -(CH 2 )4-NH 2 and - (CH2)3-NH-C(NH2)-NH2.
  • Rio also includes l-guanidinoprop-3-yl, benzyl, 4- hydroxybenzyl, imidazol-4-yl, indol-3-yl, methoxyphenyl and ethoxyphenyl.
  • Another set of protecting groups include the residue of an amino- containing compound, in particular an amino acid, a polypeptide, a protecting group, -N ⁇ SO2R NHC(O)R, -N(R)2, NH2 or -NH(R)(H), whereby for example a carboxylic acid is reacted, i.e. coupled, with the amine to form an amide, as in C(O)NR 2 .
  • a phosphonic acid may be reacted with the amine to form a phosphonamidate, as in -P(O)(OR)(NR 2 ).
  • amino acids have the structure R 17 C(O)CH(R 16 )NH-, where
  • R is -OH, -OR, an amino acid or a polypeptide residue.
  • Amino acids are low molecular weight compounds, on the order of less than about 1000 MW and which contain at least one amino or imino group and at least one carboxyl group. Generally the amino acids will be found in nature, i.e., can be detected in biological material such as bacteria or other microbes, plants, animals or man. Suitable amino acids typically are alpha amino acids, i.e. compounds characterized by one amino or imino nitrogen atom separated from the carbon atom of one carboxyl group by a single substituted or unsubstituted alpha carbon atom.
  • hydrophobic residues such as mono-or di-alkyl or aryl amino acids, cycloalkylamino acids and the like. These residues contribute to cell permeability by increasing the partition coefficient ofthe parental drug. Typically, the residue does not contain a sulfhydryl or guanidino substituent.
  • Naturally-occurring amino acid residues are those residues found naturally in plants, animals or microbes, especially proteins thereof. Polypeptides most typically will be substantially composed of such naturally- occurring amino acid residues.
  • amino acids are glycine, alanine, valine, leucine, isoleucine, serine, threonine, cysteine, methionine, glutamic acid, aspartic acid, Iysine, hydroxylysine, arginine, histidine, phenylalanine, tyrosine, tryptophan, proline, asparagine, glutamine and hydroxyproline.
  • unnatural amino acids for example, valanine, phenylglycine and homoarginine are also included.
  • Commonly encountered amino acids that are not gene- encoded may also be used in the present invention. All ofthe amino acids used in the present invention may be either the D- or L- optical isomer.
  • any site in the parental molecule is amidated with an amino acid as described herein, although it is within the scope of this invention to introduce amino acids at more than one permitted site.
  • a carboxyl group of R 3 is amidated with an amino acid.
  • the ⁇ -amino or ⁇ -carboxyl group ofthe amino acid or the terminal amino or carboxyl group of a polypeptide are bonded to the parental functionalities, i.e., carboxyl or amino groups in the amino acid side chains generally are not used to form the amide bonds with the parental compound (although these groups may need to be protected during synthesis ofthe conjugates as described further below).
  • carboxyl-containing side chains of amino acids or polypeptides it will be understood that the carboxyl group optionally will be blocked, e.g., by R 1 , esterified with R 5 or amidated. Similarly, the amino side chains R 16 optionally will be blocked with R 1 or substituted with R 5 .
  • Such ester or amide bonds with side chain amino or carboxyl groups like the esters or amides with the parental molecule, optionally are hydrolyzable in vivo or in vitro under acidic (pH ⁇ 3) or basic (pH >10) conditions.
  • esters or amino acid or polypeptide amidates also are useful as intermediates for the preparation ofthe parental molecule containing free amino or carboxyl groups.
  • the free acid or base ofthe parental compound for example, is readily formed from the esters or amino acid or polypeptide conjugates of this invention by conventional hydrolysis procedures.
  • an amino acid residue contains one or more chiral centers, any of the D, L, meso, threo or erythro (as appropriate) racemates, scalemates or mixtures thereof may be used.
  • D isomers are useful.
  • L isomers are more versatile since they can be susceptible to both non-enzymatic and enzymatic hydrolysis, and are more efficiently transported by amino acid or dipeptidyl transport systems in the gastrointestinal tract.
  • suitable amino acids whose residues are represented by R x or R y include the following: Glycine; Aminopolycarboxylic acids, e.g., aspartic acid, ⁇ -hydroxyaspartic acid, glutamic acid, ⁇ -hydroxyglutamic acid, ⁇ -methylaspartic acid, ⁇ -methylglutamic acid, ⁇ , ⁇ -dimethylaspartic acid, ⁇ -hydroxyglutamic acid, ⁇ , ⁇ -dihydroxyglutamic acid, ⁇ -phenylglutamic acid, ⁇ -methyleneglutamic acid, 3-aminoadipic acid, 2- aminopimelic acid, 2-aminosuberic acid and 2-aminosebacic acid; Amino acid amides such as glutamine and asparagine; Polyamino- or polybasic-monocarboxylic acids such as arginine, Iysine, ⁇ -aminoalanine, ⁇ -aminobutyrine, ornith
  • Polypeptides are polymers of amino acids in which a carboxyl group of one amino acid monomer is bonded to an amino or imino group ofthe next amino acid monomer by an amide bond.
  • Polypeptides include dipeptides, low molecular weight polypeptides (about 1500-5000 MW) and proteins. Proteins optionally contain 3, 5, 10, 50, 75, 100 or more residues, and suitably are substantially sequence-homologous with human, animal, plant or microbial proteins. They include enzymes (e.g., hydrogen peroxidase) as well as immunogens such as KLH, or antibodies or proteins of any type against which one wishes to raise an immune response. The nature and identity ofthe polypeptide may vary widely.
  • the polypeptide amidates are useful as immunogens in raising antibodies against either the polypeptide (if it is not immunogenic in the animal to which it is administered) or against the epitopes on the remainder ofthe compound of this invention.
  • Antibodies capable of binding to the parental non-peptidyl compound are used to separate the parental compound from mixtures, for example in diagnosis or manufacturing ofthe parental compound.
  • the conjugates of parental compound and polypeptide generally are more immunogenic than the polypeptides in closely homologous animals, and therefore make the polypeptide more immunogenic for facilitating raising antibodies against it.
  • the polypeptide or protein may not need to be immunogenic in an animal typically used to raise antibodies, e.g., rabbit, mouse, horse, or rat, but the final product conjugate should be immunogenic in at least one of such animals.
  • the polypeptide optionally contains a peptidolytic enzyme cleavage site at the peptide bond between the first and second residues adjacent to the acidic heteroatom. Such cleavage sites are flanked by enzymatic recognition structures, e.g., a particular sequence of residues recognized by a peptidolytic enzyme.
  • Peptidolytic enzymes for cleaving the polypeptide conjugates of this invention are well known, and in particular include carboxypeptidases.
  • Carboxypeptidases digest polypeptides by removing C-terminal residues, and are specific in many instances for particular C-terminal sequences. Such enzymes and their substrate requirements in general are well known.
  • a dipeptide (having a given pair of residues and a free carboxyl terminus) is covalently bonded through its ⁇ -amino group to the phosphorus or carbon atoms ofthe compounds herein.
  • Wi is phosphonate it is expected that this peptide will be cleaved by the appropriate peptidolytic enzyme, leaving the carboxyl ofthe proximal amino acid residue to autocatalytically cleave the phosphonoamidate bond.
  • Suitable dipeptidyl groups are AA, AR, AN, AD, AC, AE, AQ, AG, AH, AI, AL, AK, AM, AF, AP, AS, AT, AW, AY, AV, RA, RR, RN, RD, RC, RE, RQ, RG, RH, Rl, RL, RK, RM, RF, RP, RS, RT, RW, RY, RV, NA, NR, NN, ND, NC, NE, NQ, NG, NH, NI, NL, NK, NM, NF, NP, NS, NT, NW, NY, NV, DA, DR, DN, DD, DC, DE, DQ, DG, DH, DI, DL, DK, DM, DF, DP, DS, DT, DW, DY, DV, CA, CR, CN, CD, CC, CE, C
  • Tripeptide residues are also useful as protecting groups.
  • the sequence -X 4 -pro-X 5 - (where X 4 is any amino acid residue and X 5 is an amino acid residue, a carboxyl ester of proline, or hydrogen) will be cleaved by luminal carboxypeptidase to yield X 4 with a free carboxyl, which in turn is expected to autocatalytically cleave the phosphonoamidate bond.
  • the carboxy group of X 5 optionally is esterified with benzyl.
  • Dipeptide or tripeptide species can be selected on the basis of known transport properties and/or susceptibility to peptidases that can affect transport to intestinal mucosal or other cell types.
  • Dipeptides and tripeptides lacking an ⁇ - amino group are transport substrates for the peptide transporter found in brush border membrane of intestinal mucosal cells (Bai, J.P.F., (1992) Pharm Res. 9:969-978).
  • Transport competent peptides can thus be used to enhance bioavailability ofthe amidate compounds.
  • Di- or tripeptides having one or more amino acids in the D configuration are also compatible with peptide transport and can be utilized in the amidate compounds of this invention. Amino acids in the D configuration can be used to reduce the susceptibility of a di- or tripeptide to hydrolysis by proteases common to the brush border such as aminopeptidase N.
  • di- or tripeptides alternatively are selected on the basis of their relative resistance to hydrolysis by proteases found in the lumen ofthe intestine.
  • tripeptides or polypeptides lacking asp and/or glu are poor substrates for aminopeptidase A
  • di- or tripeptides lacking amino acid residues on the N-terminal side of hydrophobic amino acids are poor substrates for endopeptidase
  • peptides lacking a pro residue at the penultimate position at a free carboxyl terminus are poor substrates for carboxypeptidase P.
  • a 1 is of the formula:
  • a 1 is of the formula:
  • a 1 is of the formula:
  • a 1 is of the formula:
  • W ⁇ -5a is a carbocycle or a heterocycle where W 5a a . is independently substituted with 0 or 1 R groups.
  • a specific value for Ml 2a is 1.
  • a 1 is ofthe formula:
  • a 1 is of the formula:
  • W 5a is a carbocycle independently substituted with 0 or 1 R 2 groups;
  • a 1 is ofthe formula:
  • a 1 is ofthe formula:
  • W 5a is a carbocycle independently substituted with 0 or 1 R 2 groups;
  • a 1 is ofthe formula:
  • W 5a is a carbocycle or heterocycle where W 5a is independently substituted with 0 or 1 R 2 groups.
  • a 1 is ofthe formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • Y 2b is O or N(R X ); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8.
  • Ml 2d is 1.
  • a 3 is ofthe formula:
  • a 3 is of the formula:
  • W 5 is a carbocycle.
  • a 3 is of the formula:
  • W 5 is phenyl.
  • a 3 is of the formula:
  • a 3 is of the formula:
  • Y 2b is O or N(R X ); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8.
  • R 1 is H.
  • a 3 is ofthe formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • a 3 is ofthe formula:
  • a 3 is ofthe formula:
  • a 3 is ofthe formula:
  • a 3 is of the formula:
  • a 3 is of the formula:
  • Y la is O or S
  • Y 2b is O or N(R 2 );
  • Y 2c is O, N(R y ) or S.
  • a 3 is of the formula:
  • a 3 is ofthe formula:
  • a 3 is ofthe formula:
  • Y 2 is O or N(R 2 ).
  • Y 2b is O or N(R X ); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8.
  • a 3 is ofthe formula:
  • a 3 is of the formula:
  • a 0 is of the formula:
  • each R is independently (C 1 -C 6 )alkyl.
  • R x is independently H, R , W , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group;
  • R 1 is independently H or alkyl of 1 to 18 carbon atoms;
  • R 2 is independently H, R 1 , R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R 3 groups or taken together at a carbon atom, two R 2 groups form a ring of 3 to 8 carbons and the ring maybe substituted with 0 to 3 R 3 groups;
  • R x is ofthe formula:
  • R x is ofthe formula:
  • R x is ofthe formula:
  • R y is hydrogen or alkyl of 1 to
  • R x is ofthe formula:
  • Y 1 is O or S
  • Y 2 is O, N(R y ) or S.
  • R x is a group ofthe formula:
  • mla, mlb, mlc, mid and mle are independently 0 or 1; ml2c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; R y is H, W 3 , R 2 or a protecting group; provided that: if mla, ml2c, and mid are 0, then mlb, mlc and mle are 0; if mla and ml2c are 0 and mid is not 0, then mlb and mlc are 0; if mla and mid are 0 and ml2c is not 0, then mlb and at least one of mlc and mle are 0; if mla is 0 and ml2c and mid are not 0, then mlb is 0; if ml2c and mid are 0 and mla is not 0, then at least two of mlb, mlc and mle are 0; if ml2c and mid
  • W 5 carbocycles and W heterocycles may be independently substituted with 0 to 3 R 2 groups.
  • W 5 may be a saturated, unsaturated or aromatic ring comprising a mono- or bicyclic carbocycle or heterocycle.
  • W 5 may have 3 to 10 ring atoms, e.g., 3 to 7 ring atoms.
  • the W 5 rings are saturated when containing 3 ring atoms, saturated or mono-unsaturated when containing 4 ring atoms, saturated, or mono- or di-unsaturated when containing 5 ring atoms, and saturated, mono- or di-unsaturated, or aromatic when containing 6 ring atoms.
  • a W 5 heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S).
  • W 5 heterocyclic monocycles may have 3 to 6 ring atoms (2 to 5 carbon atoms and 1 to 2 heteroatoms selected from N, O, and S); or 5 or 6 ring atoms (3 to 5 carbon atoms and 1 to 2 heteroatoms selected from N and S).
  • W 5 heterocyclic bicycles have 7 to 10 ring atoms (6 to 9 carbon atoms and 1 to 2 heteroatoms selected from N, O, and S) arranged as a bicyclo [4,5], [5,5], [5,6], or [6,6] system; or 9 to 10 ring atoms (8 to 9 carbon atoms and 1 to 2 hetero atoms selected from N and S) arranged as a bicyclo [5,6] or [6,6] system.
  • the W 5 heterocycle may be bonded to Y 2 through a carbon, nitrogen, sulfur or other atom by a stable covalent bond.
  • W 5 heterocycles include for example, pyridyl, dihydropyridyl isomers, piperidine, pyridazinyl, pyrimidinyl, pyrazinyl, s-triazinyl, oxazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, furanyl, thiofuranyl, thienyl, and pyrrolyl.
  • W 5 also includes, but is not limited to, examples such as:
  • W 5 carbocycles and heterocycles may be independently substituted with 0 to 3 R 2 groups, as defined above.
  • substituted W 5 carbocycles include:
  • substituted phenyl carbocycles include:
  • the invention provides conjugates that comprise a kinase inhibiting compound that is linked to one or more phosphonate groups either directly (e.g. through a covalent bond) or through a linking group (i.e. a linker).
  • a linking group i.e. a linker.
  • the nature of the linker is not critical provided it does not interfere with the ability ofthe phosphonate containing compound to function as a therapeutic agent.
  • the phosphonate or the linker can be linked to the compound (e.g. a compound of 100-103) at any synthetically feasible position on the compound by removing a hydrogen or any portion ofthe compound to provide an open valence for attachment ofthe phosphonate or the linker.
  • the linking group or linker (which ⁇ 1 can be designated "L") can include all or a portions ofthe group A , A , A , or W 3 described herein.
  • the linking group or linker has a molecular weight of from about 20 daltons to about 400 daltons.
  • the linking group or linker has a length of about 5 angstroms to about 300 angstroms.
  • the linking group or linker is a divalent, branched or unbranched, saturated or unsaturated, hydrocarbon chain, having from 2 to 25 carbon atoms, wherein one or more (e.g. 1, 2, 3, or 4) ofthe carbon atoms is optionally replaced by (-O-), and wherein the chain is optionally substituted on carbon with one or more (e.g.
  • the linking group or linker is a divalent radical formed from a peptide. In another embodiment ofthe invention the linking group or linker is a divalent radical formed from an amino acid. In another embodiment ofthe invention the linking group or linker is a divalent radical formed from poly-L-glutamic acid, poly-L-aspartic acid, poly-L- histidine, poly-L-ornithine, poly-L-serine, poly-L-threonine, poly-L-tyrosine, poly-L-leucine, poly-L-lysine-L-phenylalanine, poly-L-lysine or poly-L-lysine- L-tyrosine.
  • the linking group or linker is methylene, ethylene, or propylene.
  • the linking group or linker is attached to the phosphonate group through a carbon atom ofthe linker.
  • Intracellular Targeting The phosphonate group ofthe compounds ofthe invention may cleave in vivo in stages after they have reached the desired site of action, i.e. inside a cell.
  • One mechanism of action inside a cell may entail a first cleavage, e.g. by esterase, to provide a negatively-charged "locked-in" intermediate. Cleavage of a terminal ester grouping in a compound ofthe invention thus affords an unstable intermediate which releases a negatively charged "locked in” intermediate.
  • intracellular enzymatic cleavage or modification ofthe phosphonate or prodrug compound may result in an intracellular accumulation ofthe cleaved or modified compound by a "trapping" mechanism.
  • the cleaved or modified compound may then be "locked-in” the cell by a significant change in charge, polarity, or other physical property change which decreases the rate at which the cleaved or modified compound can exit the cell, relative to the rate at which it entered as the phosphonate prodrug.
  • Other mechanisms by which a therapeutic effect are achieved may be operative as well.
  • Enzymes which are capable of an enzymatic activation mechanism with the phosphonate prodrug compounds ofthe invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphatases. From the foregoing, it will be apparent that many different drugs can be derivatized in accord with the present invention. Numerous such drugs are specifically mentioned herein. However, it should be understood that the discussion of drug families and their specific members for derivatization according to this invention is not intended to be exhaustive, but merely illustrative.
  • Kinase-inhibitory Compounds The compounds ofthe invention include those with kinase-inhibitory activity.
  • the compounds ofthe inventions bear one or more (e.g.
  • kinase-inhibitory compound includes those compounds that inhibit the activity of at least one kinase.
  • the compounds include CP-690,550, AP23464, A-420983 and roscovitine.
  • compounds ofthe invention have a molecular weight of from about 400 amu to about 10,000 amu; in a specific embodiment ofthe invention, compounds have a molecular weight of less than about 5000 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 2500 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 1000 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 800 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 600 amu; and in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 600 amu and a molecular weight of greater than about 400 amu.
  • the compounds ofthe invention also typically have a logD (polarity) less than about 5.
  • the invention provides compounds having a logD less than about 4; in another embodiment the invention provides compounds having a logD less than about 3; in another embodiment the invention provides compounds having a logD greater than about -5; in another embodiment the invention provides compounds having a logD greater than about -3; and in another embodiment the invention provides compounds having a logD greater than about 0 and less than about 3.
  • Selected substituents within the compounds ofthe invention are present to a recursive degree.
  • "recursive substituent" means that a substituent may recite another instance of itself. Because ofthe recursive nature of such substituents, theoretically, a large number may be present in any given embodiment.
  • R x contains a R y substituent.
  • R y can be R 2 , which in turn can be R 3 . If R 3 is selected to be R 3 °, then a second instance of R x can be selected.
  • R y is R 2 , which in turn can be R 3 . If R 3 is selected to be R 3 °, then a second instance of R x can be selected.
  • properties include, by way of example and not limitation, physical properties such as molecular weight, solubility or log P, application properties such as activity against the intended target, and practical properties such as ease of synthesis.
  • W 3 , R y and R 3 are all recursive substituents in certain embodiments.
  • each of these may independently occur 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or 0, times in a given embodiment. More typically, each of these may independently occur 12 or fewer times in a given embodiment. More typically yet, W 3 will occur 0 to 8 times, R y will occur 0 to 6 times and R 3 will occur 0 to 10 times in a given embodiment. Even more typically, W 3 will occur 0 to 6 times, R y will occur 0 to 4 times and R 3 will occur 0 to 8 times in a given embodiment.
  • Recursive substituents are an intended aspect ofthe invention. One of ordinary skill in the art of medicinal chemistry understands the versatility of such substituents.
  • the total number will be determined as set forth above.
  • a compound described herein is substituted with more than one ofthe same designated group, e.g., "R 1 " or “R 6a “ 5 then it will be understood that the groups may be the same or different, i.e., each group is independently selected. Wavy lines indicate the site of covalent bond attachments to the adjoining groups, moieties, or atoms.
  • the compound is in an isolated and purified form.
  • isolated and purified means that the compound is substantially free from biological materials (e.g. blood, tissue, cells, etc.).
  • the term means that the compound or conjugate ofthe invention is at least about 50 wt.% free from biological materials; in another specific embodiment, the term means that the compound or conjugate ofthe invention is at least about 75 wt.% free from biological materials; in another specific embodiment, the term means that the compound or conjugate ofthe invention is at least about 90 wt.% free from biological materials; in another specific embodiment, the term means that the compound or conjugate ofthe invention is at least about 98 wt.% free from biological materials; and in another embodiment, the term means that the compound or conjugate ofthe invention is at least about 99 wt.% free from biological materials.
  • the invention provides a compound or conjugate ofthe invention that has been synthetically prepared (e.g., ex vivo).
  • the compound is not an anti- inflammatory compound; in another embodiment the compound is not an anti- infective; in another embodiment the compound is not a compound that is active against immune-mediated conditions; in another embodiment the compound is not a compound that is active against metabolic diseases; in another embodiment the compound is not an antiviral agent; in another embodiment the compound is not a nucleoside; in another embodiment the compound is not a IMPDH inhibitor; in another embodiment the compound is not an antimetabolite; in another embodiment the compound is not a PNP inhibitor; in another embodiment the compound inhibits a serine/threonine kinase, tyrosine kinase, Bcr-Abl kinase, cyclin-dependent kinase, Flt3 tyrosine kinase, MAP Erk kinase, JAK3 kina
  • the invention provides a conjugate comprising a kinase inhibiting compound linked to one or more phosphonate groups; or a pharmaceutically acceptable salt or solvate thereof, wherein the kinase inhibiting compound is not Gefitinib, imatinib, erlotinib, vatalanib, alvocidib, CEP-701, GLEEVEC, midostaurin, MLN-518, PD-184352, doramapimod, BAY-43-9006, or CP-690,550.
  • the invention provides a compound of any one of formulae 500-511:
  • A is A , A 2 or W 3 with the proviso that the conjugate includes at least one A A 1 is:
  • a 2 is:
  • a 3 is: Y 1 is independently O, S, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), or N(N(R X )( R X )); Y 2 is independently a bond, O, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), N(N(R X )( R x )), -S(O)M2-, or -S(O) M 2-S(O) M 2-; and when Y 2 joins two 9 9 9 phosphorous atoms Y can also be C(R )(R ); R x is independently H, R 1 , R 2 , W 3 , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group
  • R 1 is independently H or alkyl of 1 to 18 carbon atoms
  • R 2 is independently H, R 1 , R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R groups or taken together at a carbon atom, two R groups form a ring of 3 to 8 carbons and the ring may be substituted with 0 to 3 R 3 groups
  • R 3 is R 3a , R 3b , R 3c or R 3d , provided that when R 3 is bound to a heteroatom, then R 3 is R 3c or R 3d
  • R 3a is F, Cl, Br, I, -CN, N 3 or -NO 2
  • R 3b is Y 1
  • R 3c is -R x , -N(R X )(R X ), -SR X , -S(O)R x , -S(O) 2 R x , -S(
  • R 3d is -C(Y 1 )R X , -C(Y * -)OR x or -C(Y J )(N(R X )(R X ));
  • R 4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms;
  • R 5 is R 4 wherein each R 4 is substituted with 0 to 3 R 3 groups;
  • W 3 is W 4 or W 5 ;
  • W 4 is R 5 , -C(Y 2 )R 5 , -CC ⁇ W 5 , -SO M2
  • the invention provides a kinase inhibiting conjugate that excludes such a compound.
  • the invention provides a compound ofthe formula: [DRUG]-(A°) nn or a pharmaceutically acceptable salt or solvate thereof wherein, DRUG is a compound of any one of formulae 500-511 (illustrated above); nn is 1, 2, or 3; A 0 is A 1 , A 2 or W 3 with the proviso that the conjugate includes at least one A 1 ; A 1 is:
  • a 2 is:
  • a 3 is:
  • Y 1 is independently O, S, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), or N(N(R X )( R X ));
  • Y 2 is independently a bond, O, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), N(N(R X )( R x )), -S(O)M2-, or -S(O) M 2-S(O) M 2-; and when Y 2 joins two phosphorous atoms Y 2 can also be C(R 2 )(R 2 );
  • R x is independently H, R 1 , R 2 , W 3 , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group;
  • R 1 is independently H or alkyl of 1 to 18 carbon atoms;
  • R 2 is independently H, R 1 , R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R 3 groups or taken together at a carbon atom, two R 2 groups form a ring of 3 to 8 carbons and the ring may be substituted with 0 to 3 R 3 groups;
  • R 3 is R 3a , R 3b , R 3c or R 3d , provided that when R 3 is bound to a heteroatom, then R 3 is R 3 ° or R 3d ;
  • R 3a is F, Cl, Br, I, -CN, N 3 or -NO 2 ;
  • R 3b is Y 1 ;
  • R 30 is -R x , -N(R X )(R X ), -SR X , -S(O)R x , -S(O
  • R 3d is -C ⁇ R", -C(Y 1 )OR x or -C(Y 1 )(N(R X )(R X ));
  • R 4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of
  • W 5 is carbocycle or heterocycle wherein W 5 is independently substituted with 0 to 3 R 2 groups; W is W independently substituted with 1, 2, or 3 A groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; and M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the invention provides a kinase inhibiting conjugate that excludes such a compound.
  • the invention provides a compound of any one of formulae 1-36:
  • a 0 is A 1 ;
  • a 1 is:
  • a 3 is:
  • Y 1 is independently O, S, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), or N(N(R X )( R X ));
  • Y 2 is independently a bond, O, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), N(N(R X )( R x )), -S(O) M 2-, or -S(O) M2 -S(O) M 2-; and when Y 2 joins two phosphorous atoms Y 2 can also be C(R 2 )(R 2 );
  • R x is independently H, R 2 , W 3 , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group;
  • R 1 is independently H or alkyl of 1 to 18 carbon atoms;
  • R 2 is independently H, R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R groups;
  • R J is R ,3a , r R,3b , R ,3 i c C or R ,3d , provided that when R' is bound to a heteroatom, then R 3 is R 3c or R 3d ;
  • R 3a is F, Cl, Br, I, -CN, N 3 or -NO 2 ;
  • R 3 is Y 1 ;
  • R 3c is -R x , -N(R X )(R X ), -SR X , -S(O)R x , -S(O) 2 R x , -S(O)(OR x ), - S(O) 2 (OR x ), -
  • R 4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms
  • R 5 is R 4 wherein each R 4 is substituted with 0 to 3 R 3 groups
  • R 5a is independently alkylene of 1 to 18 carbon atoms, alkenylene of 2 to 18 carbon atoms, or alkynylene of 2-18 carbon atoms any one of which alkylene, alkenylene or alkynylene is substituted with 0-3 R 3 groups
  • W 3 is W 4 or W 5 ;
  • W 4 is R 5 , -CO W 5 , -SO 2 R 5 , or -SO 2 W 5 ;
  • W 5 is carbocycle or heterocycle wherein W 5 is independently substituted with 0 to 3 R 2 groups;
  • W 6 is W 3 independently substituted with 1, 2, or 3 A 3 groups;
  • M2 is 0,
  • the invention provides a compound of any one of formulae 500a-511a:
  • a 0 is A 1 , A 2 or W 3 with the proviso that the conjugate includes at least one A 1 ; is:
  • a 2 is:
  • a 3 is: Y 1 is independently O, S, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), or N(N(R X )( R X )); Y 2 is independently a bond, O, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), N(N(R X )( R x )), -S(O)M2-, or -S(O) M2 -S(O) M 2-; and when Y 2 joins two phosphorous atoms Y 2 can also be C(R 2 )(R 2 ); R x is independently H, R 1 , R 2 , W 3 , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group;
  • R 1 is independently H or alkyl of 1 to 18 carbon atoms;
  • R 2 is independently H, R 1 , R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R groups or taken together at a carbon atom, two R groups form a ring of 3 to 8 carbons and the ring may be substituted with 0 to 3 R 3 groups;
  • R 3 is R 3a , R 3 , R 3c or R 3d , provided that when R 3 is bound to a heteroatom, then R 3 is R 3c or R 3d ;
  • R 3a is F, Cl, Br, I, -CN, N 3 or -NO 2 ;
  • R ⁇ is Y 1 ;
  • R 3c is -R x , -N(R X )(R X ), -SR X , -S(O)R x , -S(O) 2 R
  • a 0 is A 1 ;
  • a 1 is:
  • a 3 is:
  • Y 1 is independently O, S, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), or N(N(R X )( R x ));
  • Y 2 is independently a bond, O, N(R X ), N(O)(R x ), N(OR x ), N(O)(OR x ), N(N(R X )( R x )), -S(O)M2-, or -S(O) M 2-S(O) M 2-; and when Y 2 joins two 9 9 9 phosphorous atoms Y can also be C(R )(R );
  • R x is independently H, R 2 , W 3 , a protecting group, or the formula:
  • R y is independently H, W 3 , R 2 or a protecting group;
  • R 1 is independently H or alkyl of 1 to 18 carbon atoms;
  • R 2 is independently H, R 3 or R 4 wherein each R 4 is independently substituted with 0 to 3 R 3 groups;
  • R 3 is R 3a , R 3b , R 3c or R 3d , provided that when R 3 is bound to a heteroatom, then R 3 is R 3c or R 3d ;
  • R 3a is F, Cl, Br, I, -CN, N 3 or -NO 2 ;
  • R 3b is Y !
  • R 3c is -R x , -N(R X )(R X ), -SR X , -S(O)R x , -S(O) 2 R x , -S(O)(OR x ), -
  • R 3d is -C ⁇ R", -C(Y J )OR x or -C(Y 1 )(N(R X )(R X ));
  • R 4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl
  • the invention is provides compounds capable of accumulating in human PBMC (peripheral blood mononuclear cells).
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • PBMC are critical components ofthe mechanism against infection.
  • PBMC may be isolated from heparinized whole blood of normal healthy donors or buffy coats, by standard density gradient centrifugation and harvested from the interface, washed (e.g. phosphate-buffered saline) and stored in freezing medium.
  • PBMC may be cultured in multi-well plates. At various times of culture, supernatant may be either removed for assessment, or cells may be harvested and analyzed (Smith R. etal (2003) Blood 102(7):2532-2540).
  • the compounds of this embodiment may further comprise a phosphonate or phosphonate prodrug. More typically, the phosphonate or phosphonate prodrug can have the structure A 3 as described herein.
  • compounds ofthe invention demonstrate improved intracellular half-life ofthe compounds or intracellular metabolites ofthe compounds in human PBMC when compared to analogs ofthe compounds not having the phosphonate or phosphonate prodrug.
  • the half-life is improved by at least about 50%, more typically at least in the range 50-100%, still more typically at least about 100%, more typically yet greater than about 100%.
  • the intracellular half-life of a metabolite ofthe compound in human PBMCs is improved when compared to an analog ofthe compound not having the phosphonate or phosphonate prodrug.
  • the metabolite may be generated intracellularly, e.g. generated within human PBMC.
  • the metabolite may be a product ofthe cleavage of a phosphonate prodrug within human PBMCs.
  • the phosphonate prodrug may be cleaved to form a metabolite having at least one negative charge at physiological pH.
  • the phosphonate prodrug may be enzymatically cleaved within human PBMC to form a phosphonate having at least one active hydrogen atom of the form P-OH.
  • Stereoisomers The compounds ofthe invention may have chiral centers, e.g. , chiral carbon or phosphorus atoms.
  • the compounds ofthe invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers, and atropisomers.
  • the compounds ofthe invention include enriched or resolved optical isomers at any or all asymmetric, chiral atoms. In other words, the chiral centers apparent from the depictions are provided as the chiral isomers or racemic mixtures.
  • racemic and diastereomeric mixtures are separated into their individual, substantially optically pure isomers through well-known techniques such as, for example, the separation of diastereomeric salts formed with optically active adjuncts, e.g., acids or bases followed by conversion back to the optically active substances.
  • optically active adjuncts e.g., acids or bases followed by conversion back to the optically active substances.
  • the desired optical isomer is synthesized by means of stereospecific reactions, beginning with the appropriate stereoisomer ofthe desired starting material.
  • the compounds ofthe invention can also exist as tautomeric isomers in certain cases.
  • compositions of this invention optionally comprise salts ofthe compounds herein, especially pharmaceutically acceptable non-toxic salts containing, for example, Na + , Li + , K + > Ca + 2 and Mg + 2.
  • Such salts may include those derived by combination of appropriate cations such as alkali and alkaline earth metal ions or ammonium and quaternary amino ions with an acid anion moiety, typically a carboxylic acid.
  • Monovalent salts are preferred if a water soluble salt is desired.
  • Metal salts typically are prepared by reacting the metal hydroxide with a compound of this invention. Examples of metal salts which are prepared in this way are salts containing Li + , Na + , and K + . A less soluble metal salt can be precipitated from the solution of a more soluble salt by addition ofthe suitable metal compound.
  • salts may be formed from acid addition of certain organic and inorganic acids, e.g., HC1, HBr, H2SO4 5 H3PO4 or organic sulfonic acids, to basic centers, typically amines, or to acidic groups.
  • organic and inorganic acids e.g., HC1, HBr, H2SO4 5 H3PO4 or organic sulfonic acids
  • the compositions herein comprise compounds ofthe invention in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
  • the salts ofthe parental compounds with one or more amino acids are also included within the scope of this invention.
  • any ofthe amino acids described above are suitable, especially the naturally-occurring amino acids found as protein components, although the amino acid typically is one bearing a side chain with a basic or acidic group, e.g., Iysine, arginine or glutamic acid, or a neutral group such as glycine, serine, threonine, alanine, isoleucine, or leucine.
  • a basic or acidic group e.g., Iysine, arginine or glutamic acid
  • a neutral group such as glycine, serine, threonine, alanine, isoleucine, or leucine.
  • compositions ofthe invention may act as kinase inhibitors, as intermediates for such inhibitors, or have other utilities as described herein.
  • the inhibitors will bind to at least one kinase.
  • Compositions binding the kinase may bind with varying degrees of reversibility. Those compounds binding substantially irreversibly are ideal candidates for use in this method ofthe invention. Once labeled, the substantially irreversibly binding compositions are useful as probes for the detection of a kinase.
  • the invention relates to methods of detecting at least one kinase in a sample suspected of containing a kinase including the steps of: treating a sample suspected of containing kinase with a composition including a compound ofthe invention bound to a label; and observing the effect ofthe sample on the activity ofthe label.
  • Suitable labels are well known in the diagnostics field and include stable free radicals, fluorophores, radioisotopes, enzymes, chemiluminescent groups and chromogens.
  • the compounds herein are labeled in conventional fashion using functional groups such as hydroxyl or amino.
  • samples suspected of containing at least one kinase include natural or man-made materials such as living organisms; tissue or cell cultures; biological samples such as biological material samples (blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like.
  • biological material samples blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like
  • laboratory samples food, water, or air samples
  • bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like.
  • sample will be suspected of containing a kinase.
  • Samples can be contained in any medium including water and organic solvent/water mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures.
  • the treating step ofthe invention comprises adding the composition of the invention to the sample or it comprises adding a precursor ofthe composition to the sample.
  • the addition step comprises any method of administration as described above.
  • the activity of the kinase after application of the composition can be observed by any method including direct and indirect methods of detecting kinase activity. Quantitative, qualitative, and semiquantitative methods of determining kinase activity are all contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation ofthe physiological properties of a living organism are also applicable. Many organisms contain kinases.
  • the compounds of this invention are useful in the treatment or prophylaxis of conditions associated with kinase activation in animals or in man.
  • compositions ofthe invention are screened for inhibitory activity against a kinase by any ofthe conventional techniques for evaluating enzyme activity.
  • typically compositions are first screened for inhibition of kinase in vitro and compositions showing inhibitory activity are then screened for activity in vivo.
  • compositions having in vitro Ki (inhibitory constants) of less then about 5 X 10"6 M, typically less than about 1 X 10 ⁇ 7 M and preferably less than about 5 X 10"8 M are preferred for in vivo use.
  • Ki inhibitor constants
  • Useful in vitro screens have been described, e.g., Bioorg. Med. Chem.
  • compositions are formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986). Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH ofthe formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10.
  • the formulations both for veterinary and for human use, ofthe invention comprise at least one active ingredient, as above defined, together with one or more acceptable carriers therefor and optionally other therapeutic ingredients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients ofthe formulation and physiologically innocuous to the recipient thereof.
  • the formulations include those suitable for the foregoing administration routes.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any ofthe methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA).
  • Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations ofthe present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount ofthe active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be administered as a bolus, electuary or paste.
  • a tablet is made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture ofthe powdered active ingredient moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release ofthe active ingredient therefrom.
  • For administration to the eye or other external tissues e.g.
  • the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1% and 20% in increments of 0.1% w/w such as 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w.
  • the active ingredients may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredients may be formulated in a cream with an oil-in-water cream base.
  • the aqueous phase ofthe cream base may include, for example, at least 30% w/w of a polyhydric alcohol, t.e. an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3 -diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof.
  • the topical formulations may desirably include a compound which enhances absorption or penetration ofthe active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulphoxide and related analogs.
  • the oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner.
  • the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil.
  • a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat.
  • the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase ofthe cream formulations.
  • Emulgents and emulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
  • the choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties.
  • the cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers.
  • Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils are used.
  • compositions according to the present invention comprise one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents.
  • Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration.
  • tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • inert diluents such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as cellulose, microcrystalline cellulose, starch,
  • Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions ofthe invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate).
  • a suspending agent
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules ofthe invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
  • the pharmaceutical compositions ofthe invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these.
  • Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • the pharmaceutical compositions ofthe invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% ofthe total compositions (weight: weight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g ofthe active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations suitable for administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient.
  • the active ingredient is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
  • Suitable formulations include aqueous or oily solutions ofthe active ingredient.
  • Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of conditions associated with kinase activity.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition ofthe sterile liquid carrier, for example water for injection, immediately prior to use.
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets ofthe kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, ofthe active ingredient. It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefor.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
  • Compounds of the invention can also be formulated to provide controlled release ofthe active ingredient to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile ofthe active ingredient. Accordingly, the invention also provided compositions comprising one or more compounds ofthe invention formulated for sustained or controlled release. Effective dose of active ingredient depends at least on the nature ofthe condition being treated, toxicity, whether the compound is being used prophylactically (lower doses), the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day. Typically, from about 0.01 to about 10 mg/kg body weight per day. More typically, from about .01 to about 5 mg/kg body weight per day.
  • the daily candidate dose for an adult human of approximately 70 kg body weight will range from 1 mg to 1000 mg, preferably between 5 mg and 500 mg, and may take the form of single or multiple doses.
  • Routes of Administration One or more compounds ofthe invention (herein referred to as the active ingredients) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient.
  • Combination Therapy Active ingredients ofthe invention are also used in combination with other active ingredients. Such combinations are selected based on the condition to be treated, cross-reactivities of ingredients and pharmaco-properties ofthe combination. It is also possible to combine any compound ofthe invention with one or more other active ingredients in a unitary dosage form for simultaneous or sequential administration to a patient.
  • the combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
  • the combination therapy may provide "synergy” and “synergistic effect", i.e. the effect achieved when the active ingredients used together is greater than the sum ofthe effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co- formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., in separate tablets, pills or capsules, or by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
  • Metabolites ofthe Compounds ofthe Invention Also falling within the scope of this invention are the in vivo metabolic products ofthe compounds described herein.
  • the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • Such products typically are identified by preparing a radiolabelled (e.g., C 4 or j 3) compound ofthe invention, administering it parenterally in a detectable dose (e.g.
  • metabolite structures are determined in conventional fashion, e.g. , by MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art.
  • the conversion products are useful in diagnostic assays for therapeutic dosing ofthe compounds ofthe invention even if they possess no kinase inhibitory activity of their own.
  • Recipes and methods for detennining stability of compounds in surrogate gastrointestinal secretions are known. Compounds are defined herein as stable in the gastrointestinal tract where less than about 50 mole percent ofthe protected groups are deprotected in surrogate intestinal or gastric juice upon incubation for 1 hour at 37 °C. Simply because the compounds are stable to the gastrointestinal tract does not mean that they cannot be hydrolyzed in vivo.
  • the phosphonate prodrugs ofthe invention typically will be stable in the digestive system but are substantially hydrolyzed to the parental drug in the digestive lumen, liver or other metabolic organ, or within cells in general.
  • the invention also relates to methods of making the compositions ofthe invention.
  • the compositions are prepared by any ofthe applicable techniques of organic synthesis. Many such techniques are well known in the art. However, many ofthe known techniques are elaborated in Compendium of Organic
  • Work-up typically consists of quenching any unreacted reagents followed by partition between a water/organic layer system (extraction) and separating the layer containing the product.
  • Oxidation and reduction reactions are typically carried out at temperatures near room temperature (about 20 °C), although for metal hydride reductions frequently the temperature is reduced to 0 °C to -100 °C, solvents are typically aprotic for reductions and may be either protic or aprotic for oxidations. Reaction times are adjusted to achieve desired conversions.
  • Condensation reactions are typically carried out at temperatures near room temperature, although for non-equilibrating, kinetically controlled condensations reduced temperatures (0 °C to -100 °C) are also common.
  • Solvents can be either protic (common in equilibrating reactions) or aprotic (common in kinetically controlled reactions).
  • Standard synthetic techniques such as azeotropic removal of reaction by- products and use of anhydrous reaction conditions (e.g. , inert gas environments) are common in the art and will be applied when applicable.
  • the terms "treated”, “treating”, “treatment”, and the like, when used in connection with a chemical synthetic operation mean contacting, mixing, reacting, allowing to react, bringing into contact, and other terms common in the art for indicating that one or more chemical entities is treated in such a manner as to convert it to one or more other chemical entities.
  • treating compound one with compound two is synonymous with “allowing compound one to react with compound two", “contacting compound one with compound two”, “reacting compound one with compound two”, and other expressions common in the art of organic synthesis for reasonably indicating that compound one was “treated”, “reacted”, “allowed to react”, etc., with compound two. For example, treating indicates the reasonable and usual manner in which organic chemicals are allowed to react.
  • Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium, and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
  • Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like.
  • Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like.
  • the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like. Selection of appropriate methods of separation depends on the nature of the materials involved. For example, boiling point, and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply techniques most likely to achieve the desired separation.
  • a single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution ofthe racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Stereochemistry of Carbon Compounds, (1962) by E. L. Eliel, McGraw Hill; Lochmuller, C. H., (1975) J. Chromatogr., 113:(3) 283-302). Racemic mixtures of chiral compounds ofthe invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods,
  • diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, ⁇ -methyl- ⁇ -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
  • the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
  • the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. (1994) Stereochemistry of Organic Compounds. John Wiley & Sons, Inc., p. 322).
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation ofthe diastereomers and hydrolysis to yield the free, enantiomerically enriched xanthene.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, ⁇ -methoxy- ⁇ - (trifluoromethyl) ⁇ henyl acetate (Jacob III. (1982) J. Org. Chem.
  • Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (Hoye, T., WO 96/15111).
  • a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Chiral Liquid Chromatography (1989) W. J. Lough, Ed. Chapman and Hall, New York; Okamoto, (1990) J. ofChromatogr. 513:375-378).
  • Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
  • the activated precursor can be prepared by several well known methods.
  • Chlorophosphonates useful for synthesis ofthe prodrugs are prepared from the substituted- 1, 3 -propanediol (Wissner, et al, (1992) J. Med Chem. 35:1650). Chlorophosphonates are made by oxidation ofthe corresponding chlorophospholanes (Anderson, et al, (1984) J. Org. Chem. 49: 1304) which are obtained by reaction ofthe substituted diol with phosphorus trichloride. Alternatively, the chlorophosphonate agent is made by treating substituted-l,3-diols with phosphorusoxychloride (Patois, et al, (1990) J. Chem.
  • Chlorophosphonate species may also be generated in situ from corresponding cyclic phosphites (Silverburg, et al, (1996) Tetrahedron left., 31:111-11 A), which in turn can be either made from chlorophospholane or phosphoramidate intermediate.
  • Phosphoroflouridate intermediate prepared either from pyrophosphate or phosphoric acid may also act as precursor in preparation of cyclic prodrugs (Watanabe et al, (1988) Tetrahedron lett., 29:5763-66).
  • Phosphonate prodrugs ofthe present invention may also be prepared from the free acid by Mitsunobu reactions (Mitsunobu, (1981) Synthesis, 1; Campbell, (1992) J. Org. Chem. 57:6331), and other acid coupling reagents including, but not limited to, carbodiimides (Alexander, et al, (1994) Collect. Czech. Chem. Commun. 59:1853; Casara et al, (1992) Bioorg. Med. Chem. Lett.
  • aryl phosphonate esters are prepared from aryl phosphates under anionic rearrangement conditions (Melvin (1981) Tetrahedron Lett 22:3375; Casteel et al (1991) Synthesis, 691).
  • N-Alkoxy aryl salts with alkali met al derivatives of cyclic alkyl phosphonate provide general synthesis for heteroaryl-2-phosphonate linkers (Redmore (1970) J. Org. Chem. 35:4114).
  • Cyclic- 1, 3 -propanyl prodrugs of phosphonates are also synthesized from phosphonic diacids and substituted propane- 1 ,3 -diols using a coupling reagent such as 1 ,3 - dicyclohexylcarbodiimide (DCC) in presence of a base (e.g., pyridine).
  • a coupling reagent such as 1 ,3 - dicyclohexylcarbodiimide (DCC) in presence of a base (e.g., pyridine).
  • DCC dicyclohexylcarbodiimide
  • Other carbodiimide based coupling agents like 1,3-disopropylcarbodiimide or water soluble reagent, l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI) can also be utilized for the synthesis of cyclic phosphonate prodrugs.
  • the conversion of a phosphonate diester S32.1 into the corresponding phosphonate monoester S32.2 is accomplished by a number of methods.
  • the ester S32.1 in which R 1 is an aralkyl group such as benzyl is converted into the monoester compound S32.2 by reaction with a tertiary organic base such as diazabicyclooctane (DABCO) or quinuclidine, as described in J. Org * . Chem. (1995) 60:2946.
  • DABCO diazabicyclooctane
  • the reaction is performed in an inert hydrocarbon solvent such as toluene or xylene, at about 110 °C.
  • the conversion ofthe diester S32.1 in which R 1 is an aryl group such as phenyl, or an alkenyl group such as allyl, into the monoester S32.2 is effected by treatment ofthe ester S32.1 with a base such as aqueous sodium hydroxide in acetonitrile or lithium hydroxide in aqueous tetrahydrofuran.
  • Phosphonate diesters S32.1 in which one ofthe groups R 1 is aralkyl, such as benzyl, and the other is alkyl, is converted into the monoesters S32.2 in which R 1 is alkyl by hydrogenation, for example using a palladium on carbon catalyst.
  • Phosphonate diesters in which both ofthe groups R 1 are alkenyl, such as allyl, is converted into the monoester S32.2 in which R 1 is alkenyl, by treatment with chlorotris(triphenylphosphine)rhodium (Wilkinson's catalyst) in aqueous ethanol at reflux, optionally in the presence of diazabicyclooctane, for example by using the procedure described in J. Org. Chem. (1973) 38:3224, for the cleavage of allyl carboxylates.
  • the conversion of a phosphonate monoester S32.2 into a phosphonate diester S32.1 (Scheme 32, Reaction 4) in which the newly introduced R 1 group is alkyl, aralkyl, haloalkyl such as chloroethyl, or aralkyl is effected by a number of reactions in which the substrate S32.2 is reacted with a hydroxy compound R ⁇ H, in the presence of a coupling agent.
  • the second phosphonate ester group is different than the first introduced phosphonate ester group, i.e.
  • R 1 is followed by the introduction of R 2 where each of R 1 and R 2 is alkyl, aralkyl, haloalkyl such as chloroethyl, or aralkyl (Scheme 32, Reaction 4a) whereby S32.2 is converted to S32.1a.
  • Suitable coupling agents are those employed for the preparation of carboxylate esters, and include a carbodiimide such as dicyclohexylcarbodiimide, in which case the reaction is preferably conducted in a basic organic solvent such as pyridine, or (benzotriazol-1- yloxy)tripyrrolidinophosphonium hexafluorophosphate (PYBOP, Sigma), in wliich case the reaction is performed in a polar solvent such as dimethylformamide, in the presence of a tertiary organic base such as diisopropylethylamrne, or Aldrithiol-2 (Aldrich) in which case the reaction is conducted in a basic solvent such as pyridine, in the presence of a triaryl phosphine such as triphenylphosphine.
  • a carbodiimide such as dicyclohexylcarbodiimide
  • PYBOP benzotriazol-1- yloxy)
  • the conversion ofthe phosphonate monoester S32.2 to the diester S32.1 is effected by the use ofthe Mitsunobu reaction, as described above (Scheme 7).
  • the substrate is reacted with the hydroxy compound R*OH, in the presence of diethyl azodicarboxylate and a triarylphosphine such as triphenyl phosphine.
  • the phosphonate monoester S32.2 is transformed into the phosphonate diester S32.1, in which the introduced R 1 group is alkenyl or aralkyl, by reaction ofthe monoester with the halide R Br, in which R 1 is as alkenyl or aralkyl.
  • the alkylation reaction is conducted in a polar organic solvent such as dimethylformamide or acetonitrile, in the presence of a base such as cesium carbonate.
  • a polar organic solvent such as dimethylformamide or acetonitrile
  • a base such as cesium carbonate.
  • the phosphonate monoester is transformed into the phosphonate diester in a two step procedure.
  • the phosphonate monoester S32.2 is transformed into the chloro analog RP(O)(OR 1 )Cl by reaction with thionyl chloride or oxalyl chloride and the like, as described in Organic Phosphorus Compounds, G. M. Kosolapoff, L. Maeir, eds, Wiley, 1976, p.
  • Dialkyl phosphonates may be prepared according to the methods of: Quast et al (1974) Synthesis 490; Stowell et al (1990) Tetrahedron Lett. 3261; US 5663159.
  • a phosphonic acid R-link-P(O)(OH) 2 S32.3 is transformed into a phosphonate diester R-link-P(O)(OR-) S32.1 (Scheme 32, Reaction 6) by a coupling reaction with the hydroxy compound R ⁇ H, in the presence of a coupling agent such as Aldrithiol-2 (Aldrich) and triphenylphosphine.
  • the reaction is conducted in a basic solvent such as pyridine.
  • phosphonic acids S32.3 are transformed into phosphonic esters S32.1 in which R 1 is aryl, by means of a coupling reaction employing, for example, dicyclohexylcarbodiimide in pyridine at ca 70 "C.
  • phosphonic acids S32.3 are transformed into phosphonic esters S32.1 in which R 1 is alkenyl, by means of an alkylation reaction.
  • the phosphonic acid is reacted with the alkenyl bromide R*Br in a polar organic solvent such as acetonitrile solution at reflux temperature, the presence of a base such as cesium carbonate, to afford the phosphonic ester S32.1.
  • Phosphonate esters may contain a carbamate linkage.
  • the preparation of carbamates is described in Comprehensive Organic Functional Group Transformations, A. R. Katritzky, ed., Pergamon, 1995, Vol. 6, p. 416ff, and in Organic Functional Group Preparations, by S. R. Sandier and W. Karo, Academic Press, 1986, p. 260ff.
  • the carbamoyl group may be formed by reaction of a hydroxy group according to the methods known in the art, including the teachings of Ellis, US 2002/0103378 Al and Hajima, US 6018049.
  • Scheme 33 illustrates various methods by which the carbamate linkage is synthesized.
  • an alcohol S33.1 is converted into the activated derivative S33.2 in which Lv is a leaving group such as halo, imidazolyl, benztriazolyl and the like, as described herein.
  • the activated derivative S33.2 is then reacted with an amine S33.3, to afford the carbamate product S33.4.
  • Examples 1 - 7 in Scheme 33 depict methods by which the general reaction is effected.
  • Examples 8 - 10 illustrate alternative methods for the preparation of carbamates.
  • Example 1 illustrates the preparation of carbamates employing a chloroformyl derivative ofthe alcohol S33.5.
  • the alcohol S33.5 is reacted with phosgene, in an inert solvent such as toluene, at about 0 °C, as described in Org. Svn. Coll. Vol. 3, 167, 1965, or with an equivalent reagent such as trichloromethoxy chloroformate, as described in Org. Svn. Coll. Vol. 6. 715, 1988, to afford the chloroformate S33.6.
  • the latter compound is then reacted with the amine component S33.3, in the presence of an organic or inorganic base, to afford the carbamate S33.7.
  • the chloroformyl compound S33.6 is reacted with the amine S33.3 in a water- miscible solvent such as tetrahydrofuran, in the presence of aqueous sodium hydroxide, as described in Org. Syn. Coll. Vol. 3. 167, 1965, to yield the carbamate S33.7.
  • the reaction is performed in dichioromethane in the presence of an organic base such as diisopropylethylamine or dimethylaminopyridine.
  • Scheme 33 Example 2 depicts the reaction ofthe chloroformate compound S33.6 with imidazole to produce the imidazolide S33.8.
  • the imidazolide product is then reacted with the amine S33.3 to yield the carbamate S33.7.
  • the preparation ofthe imidazolide is performed in an aprotic solvent such as dichioromethane at 0°, and the preparation ofthe carbamate is conducted in a similar solvent at ambient temperature, optionally in the presence of a base such as dimethylaminopyridine, as described inJ. Med. Chem., 1989, 32, 357.
  • Scheme 33 Example 3 depicts the reaction ofthe chloroformate S33.6 with an activated hydroxyl compound R"OH, to yield the mixed carbonate ester S33.10.
  • the reaction is conducted in an inert organic solvent such as ether or dichioromethane, in the presence of a base such as dicyclohexylamine or triethylamine.
  • the hydroxyl component R"OH is selected from the group of compounds S33.19 - S33.24 shown in Scheme 33, and similar compounds.
  • the component R"OH is hydroxybenztriazole S33.19, N- hydroxysuccinimide S33.20, or pentachlorophenol, S33.21
  • the mixed carbonate S33.10 is obtained by the reaction ofthe chloroformate with the hydroxyl compound in an ethereal solvent in the presence of dicyclohexylamine, as described in Can. J. Chem., 1982, 60, 976.
  • the acyloxyimidazole S33.8 is then reacted with an equimolar amount ofthe amine R'NH 2 to afford the carbamate S33.7.
  • the reaction is performed in an aprotic organic solvent such as dichioromethane, as described in Tet. Lett, 42, 2001, 5227, to afford the carbamate S33.7.
  • Scheme 33, Example 5 illustrates the preparation of carbamates by means of an intermediate alkoxycarbonylbenztriazole S33.13. In this procedure, an alcohol ROH is reacted at ambient temperature with an equimolar amount of benztriazole carbonyl chloride S33.12, to afford the alkoxycarbonyl product S33.13.
  • the reaction is performed in an organic solvent such as benzene or toluene, in the presence of a tertiary organic amine such as triethylamine, as described in Synthesis., 1977, 704.
  • the product is then reacted with the amine R*NH 2 to afford the carbamate S33.7.
  • the reaction is conducted in toluene or ethanol, at from ambient temperature to about 80 °C as described in Synthesis., 1977, 704.
  • Scheme 33, Example 6 illustrates the preparation of carbamates in which a carbonate (R"O) 2 CO, S33.14, is reacted with an alcohol S33.5 to afford the intermediate alkyloxycarbonyl intermediate S33.15.
  • the latter reagent is then reacted with the amine R'NH 2 to afford the carbamate S33.7.
  • the procedure in which the reagent S33.15 is derived from hydroxybenztriazole S33.19 is described in Synthesis, 1993, 908; the procedure in which the reagent S33.15 is derived from N-hydroxysuccinimide S33.20 is described in Tet. Lett., 1992, 2781; the procedure in which the reagent S33.15 is derived from 2- hydroxypyridine S33.23 is described in Tet Lett, 1991, 4251; the procedure in which the reagent S33.15 is derived from 4-nitrophenol S33.24 is described in Synthesis. 1993, 103.
  • Example 7 illustrates the preparation of carbamates from alkoxycarbonyl azides S33.16.
  • an alkyl chloroformate S33.6 is reacted with an azide, for example sodium azide, to afford the alkoxycarbonyl azide S33.16.
  • the latter compound is then reacted with an equimolar amount of the amine R'NH 2 to afford the carbamate S33.7.
  • the reaction is conducted at ambient temperature in a polar aprotic solvent such as dimethylsulfoxide, for example as described in Synthesis., 1982, 404.
  • Example 8 illustrates the preparation of carbamates by means ofthe reaction between an alcohol ROH and the chloroformyl derivative of an amine S33.17.
  • the reactants are combined at ambient temperature in an aprotic solvent such as acetonitrile, in the presence of abase such as triethylamine, to afford the carbamate S33.7.
  • aprotic solvent such as acetonitrile
  • abase such as triethylamine
  • Example 10 illustrates the preparation of carbamates by means ofthe reaction between an alcohol ROH and an amine R'NH 2 .
  • an aprotic organic solvent such as tefrahydrofuran
  • a tertiary base such as triethylamine, and selenium. Carbon monoxide is passed through the solution and the reaction proceeds to afford the carbamate S33.7.
  • Activated sulfonyloxy derivatives are obtained by the reaction of phosphonic acids with trichloromethylsulfonyl chloride or with triisopropylbenzenesulfonyl chloride, as described in Tet. Lett. (1996) 7857, or Bioorg. Med. Chem. Lett. (1998) 8:663.
  • the activated sulfonyloxy derivatives are then reacted with amines or hydroxy compounds to afford amidates or esters.
  • the phosphonic acid and the amine or hydroxy reactant are combined in the presence of a diimide coupling agent.
  • the agents include Aldrithiol-2, and PYBOP and BOP, as described inJ. Org. Chem., 1995, 60, 5214, and J. Med. Chem. (1997) 40:3842, mesitylene-2-sulfonyl-3 -nitro- 1,2,4- triazole (MSNT), as described inJ. Med. Chem. (1996) 39:4958, diphenylphosphoryl azide, as described in J. Org. Chem. (1984) 49:1158, 1- (2,4,6-triisopropylbenzenesulfonyl-3-nitro-l,2,4-triazole (TPSNT) as described in Bioorg. Med. Chem. Lett.
  • Phosphonic acids are converted into amidates and esters by means ofthe Mitsunobu reaction, in which the phosphonic acid and the amine or hydroxy reactant are combined in the presence of a triaryl phosphine and a dialkyl azodicarboxylate.
  • the procedure is described in Org. Lett, 2001, 3, 643, or J. Med. Chem., 1997, 40, 3842.
  • Phosphonic esters are also obtained by the reaction between phosphonic acids and halo compounds, in the presence of a suitable base. The method is described, for example, in. Anal. Chem., 1987, 59, 1056, or J. Chem. Soc. Perkin Trans., I, 1993, 19, 2303, or J. Med.
  • Schemes 34-37 illustrate the conversion of phosphonate esters and phosphonic acids into carboalkoxy-substituted phosphonbisamidates (Scheme 34), phosphonamidates (Scheme 35), phosphonate monoesters (Scheme 36) and phosphonate diesters, (Scheme 37).
  • Scheme 38 illustrates synthesis of gem- dialkyl amino phosphonate reagents.
  • Scheme 34 illustrates various methods for the conversion of phosphonate diesters S34.1 into phosphonbisamidates S34.5.
  • the diester S34.1 prepared as described previously, is hydrolyzed, either to the monoester S34.2 or to the phosphonic acid S34.6.
  • the methods employed for these transformations are described above.
  • the monoester S34.2 is converted into the monoamidate S34.3 by reaction with an aminoester S34.9, in which the group R 2 is H or alkyl; the group R is a divalent alkylene moiety such as, for example, CHCH 3 ,
  • the group R is C 1 -C 12 alkyl, such as methyl, ethyl, propyl, isopropyl, or isobutyl; C 6 -C 20 aryl, such as phenyl or substituted phenyl; or C 6 -C 20 arylalkyl, such as benzyl or benzyhydryl.
  • a coupling agent such as a carbodiimide, for example dicyclohexyl carbodiimide, as described in J. Am. Chem.
  • amidate product S34.3 optionally in the presence of an activating agent such as hydroxybenztriazole, to yield the amidate product S34.3.
  • the amidate- forming reaction is also effected in the presence of coupling agents such as BOP, as described inJ. Org. Chem. (1995) 60:5214, Aldrithiol, PYBOP and similar coupling agents used for the preparation of amides and esters.
  • the reactants S34.2 and S34.9 are transformed into the monoamidate S34.3 by means of a Mitsunobu reaction.
  • the preparation of amidates by means ofthe Mitsunobu reaction is described inJ Med. Chem. (1995) 38:2742.
  • the benzyl group is then removed, for example by hydrogenolysis over a palladium catalyst, to give the monoacid product S34.18 which may be unstable according to J. Med. Chem. (1997) 40(23):3842.
  • This compound S34.18 is then reacted in a Mitsunobu reaction with ethyl leucinate S34.19, triphenyl phosphine and diethylazodicarboxylate, as described inJ Med. Chem., 1995, 38, 2742, to produce the bisamidate product S34.20.
  • the phosphonic acid S34.6 is converted into the mono or bis-activated derivative S34.7, in which Lv is a leaving group such as chloro, imidazolyl, triisopropylbenzenesulfonyloxy etc.
  • Lv is a leaving group such as chloro, imidazolyl, triisopropylbenzenesulfonyloxy etc.
  • the phosphonic acid is activated by reaction with triisopropylbenzenesulfonyl chloride, as described in Nucleosides and Nucleotides, 2000, 10, 1885.
  • the activated product is then reacted with the aminoester S34.9, in the presence of a base, to give the bisamidate S34.5.
  • the reaction is performed in one step, in which case the nitrogen substituents present in the product S34.5 are the same, or in two steps, via the intermediate S34.ll, in which case the nitrogen substituents can be different. Examples of these methods are shown in Scheme 34, Examples 3 and 5.
  • Example 3 a phosphonic acid S34.6 is reacted with ten molar equivalents of thionyl chloride, as described in Zh. Obschei Khim., 1958, 28, 1063, to give the dichloro compound S34.23.
  • the product is then reacted at reflux temperature in a polar aprotic solvent such as acetonitrile, and in the presence of a base such as triethylamine, with butyl serinate S34.24 to afford the bisamidate product S34.25.
  • a polar aprotic solvent such as acetonitrile
  • a base such as triethylamine
  • the intermediate monoamidate S34.3 is also prepared from the monoester S34.2 by first converting the monoester into the activated derivative S34.8 in which Lv is a leaving group such as halo, imidazolyl etc, using the procedures described above.
  • the product S34.8 is then reacted with an aminoester S34.9 in the presence of a base such as pyridine, to give an intermediate monoamidate product S34.3.
  • the latter compound is then converted, by removal ofthe R 1 group and coupling ofthe product with the aminoester S34.9, as described above, into the bisamidate S34.5.
  • An example of this procedure, in which the phosphonic acid is activated by conversion to the chloro derivative S34.26, is shown in Scheme 34, Example 4.
  • the phosphonic monobenzyl ester S34.15 is reacted, in dichioromethane, with thionyl chloride, as described in Tet. Letters., 1994, 35, 4097, to afford the phosphoryl chloride S34.26.
  • the product is then reacted in acetonitrile solution at ambient temperature with one molar equivalent of ethyl 3-amino-2-methylpropionate S34.27 to yield the monoamidate product S34.28.
  • the latter compound is hydrogenated in ethylacetate over a 5% palladium on carbon catalyst to produce the monoacid product S34.29.
  • the product is subjected to a Mitsunobu coupling procedure, with equimolar amounts of butyl alaninate S34.30, triphenyl phosphine, diethylazodicarboxylate and triethylamine in tetrahydrofuran, to give the bisamidate product S34.31.
  • a Mitsunobu coupling procedure with equimolar amounts of butyl alaninate S34.30, triphenyl phosphine, diethylazodicarboxylate and triethylamine in tetrahydrofuran, to give the bisamidate product S34.31.
  • ethyl 3-amino-2- methylpropionate S34.27 or butyl alaninate S34.30 different aminoesters S34.9, the corresponding products S34.5 are obtained.
  • the activated phosphonic acid derivative S34.7 is also converted into the bisamidate S34.5 via the diamino compound S34.
  • S34.6 may be treated with two different amino ester reagents simulataneously, i.e. S34.12 where R 4b or R 5b are different.
  • the resulting mixture of bisamidate products S34.5 may then be separable, e.g., by chromatography.
  • An example of this procedure is shown in Scheme 34, Example 6.
  • a dichlorophosphonate S34.23 is reacted with ammonia to afford the diamide S34.37.
  • the reaction is performed in aqueous, aqueous alcoholic or alcoholic solution, at reflux temperature.
  • the resulting diamino compound is then reacted with two molar equivalents of ethyl 2-bromo-3-mefhylbutyrate S34.38, in a polar organic solvent such as N-methylpyrrolidinone at ca. 150 °C, in the presence of a base such as potassium carbonate, and optionally in the presence of a catalytic amount of potassium iodide, to afford the bisamidate product S34.39.
  • a polar organic solvent such as N-methylpyrrolidinone at ca. 150 °C
  • a base such as potassium carbonate
  • a catalytic amount of potassium iodide to afford the bisamidate product S34.39.
  • a monophenyl phosphonate S35.7 is reacted with, for example, thionyl chloride, as described in J. Gen. Chem. USSR., 1983, 32, 367, to give the chloro product S35.8.
  • the product is then reacted, as described in Scheme 34, with ethyl alaninate, to yield the amidate S35.10.
  • the phosphonate monoester S34.1 is coupled, as described in Scheme 34, with an aminoester S34.9 to produce the amidate S35.1.
  • the R 1 substituent is then altered, by initial cleavage to afford the phosphonic acid S35.2.
  • the procedures for this transformation depend on the nature ofthe R 1 group, and are described above.
  • the phosphonic acid is then transformed into the ester amidate product S35.3, by reaction with the hydroxy compound R 3 OH, in which the group R 3 is aryl, heterocycle, alkyl, cycloalkyl, haloalkyl etc, using the same coupling procedures (carbodiimide, Aldrithiol-2, PYBOP, Mitsunobu reaction etc) described in Scheme 34 for the coupling of amines and phosphonic acids.
  • Scheme 34 Example 1
  • Examples of this method are shown in Scheme 35, Examples 1-3.
  • a monobenzyl phosphonate S35.ll is transformed by reaction with ethyl alaninate, using one ofthe methods described above, into the monoamidate S35.12.
  • the benzyl group is then removed by catalytic hydrogenation in ethylacetate solution over a 5% palladium on carbon catalyst, to afford the phosphonic acid amidate S35.13.
  • the product is then reacted in dichioromethane solution at ambient temperature with equimolar amounts of 1 -(dimethylaminopropyl)-3 -ethylcarbodiimide and trifluoroethanol S35.14, for example as described in Tet.
  • the activated phosphonate ester S34.8 is reacted with ammonia to yield the amidate S35.4.
  • the product is then reacted, as described in Scheme 34, with a haloester S35.5, in the presence of a base, to produce the amidate product S35.6.
  • the nature ofthe R 1 group is changed, using the procedures described above, to give the product S35.3.
  • the method is illustrated in Scheme 35, Example 4. In this sequence, the monophenyl phosphoryl chloride S35.18 is reacted, as described in Scheme 34, with ammonia, to yield the amino product S35.19.
  • Scheme 36 illustrates methods for the preparation of carboalkoxy- substituted phosphonate diesters in which one ofthe ester groups incorporates a carboalkoxy substituent.
  • a phosphonate monoester S34.1 prepared as described above, is coupled, using one ofthe methods described above, with a hydroxyester S36.1, in which the groups R 4b and R 5b are as described in Scheme 34.
  • equimolar amounts ofthe reactants are coupled in the presence of a carbodiimide such as dicyclohexyl carbodiimide, as described in Aust J. Chem., 1963, 609, optionally in the presence of dimethylaminopyridine, as described in Tet, 1999, 55, 12997.
  • the conversion of a phosphonate monoester S34.1 into a mixed diester S36.2 is also accomplished by means of a Mitsunobu coupling reaction with the hydroxyester S36.1, as described in Org. Lett, 2001, 643.
  • the reactants S34.1 and S36.1 are combined in a polar solvent such as tetrahydrofuran, in the presence of a triarylphosphine and a dialkyl azodicarboxylate, to give the mixed diester S36.2.
  • the R 1 substituent is varied by cleavage, using the methods described previously, to afford the monoacid product S36.3.
  • the latter compound is then coupled, in pyridine solution at ambient temperature, in the presence of dicyclohexyl carbodiimide, with one molar equivalent of 3 -hydroxypyridine S36.16 to yield the mixed diester S36.17.
  • a different hydroxyester S36.1 and/or a different hydroxy compound R 3 OH the corresponding products S36.4 are obtained.
  • the mixed diesters S36.2 are also obtained from the monoesters S34.1 via the intermediacy ofthe activated monoesters S36.5.
  • phosphorus pentachloride as described in J. Org: Chem., 2001 , 66, 329
  • triisopropylbenzenesulfonyl chloride in pyridine as described in Nucleosides and Nucleotides, 2000, 19, 1885, or with carbony
  • the corresponding products S36.2 are obtained.
  • the mixed phosphonate diesters are also obtained by an alternative route for incorporation ofthe R 3 O group into intermediates S36.3 in which the hydroxyester moiety is already incorporated.
  • the monoacid intermediate S36.3 is converted into the activated derivative S36.6 in which Lv is a leaving group such as chloro, imidazole, and the like, as previously described.
  • the activated intermediate is then reacted with the hydroxy compound R OH, in the presence of a base, to yield the mixed diester product S36.4.
  • the phosphonate esters S36.4 are also obtained by means of alkylation reactions performed on the monoesters S34.1.
  • the reaction between the monoacid S34.1 and the haloester S36.7 is performed in a polar solvent in the presence of a base such as diisopropylethylamine, as described in Anal. Chem., 1987, 59, 1056, or triethylamine, as described in J. Med. Chem., 1995, 38, 1372, or in a non-polar solvent such as benzene, in the presence of 18-crown-6, as described in Syn. Comm., 1995, 25, 3565.
  • the method is illustrated in Scheme 36, Example 5.
  • Scheme 37 illustrates methods for the preparation of phosphonate diesters in which both the ester substituents inco ⁇ orate carboalkoxy groups.
  • the compounds are prepared directly or indirectly from the phosphonic acids S34.6.
  • the phosphonic acid is coupled with the hydroxyester S37.2, using the conditions described previously in Schemes 34- 36, such as coupling reactions using dicyclohexyl carbodiimide or similar reagents, or under the conditions ofthe Mitsunobu reaction, to afford the diester product S37.3 in which the ester substituents are identical.
  • This method is illustrated in Scheme 37, Example 1.
  • the phosphonic acid S34.6 is reacted with three molar equivalents of butyl lactate S37.5 in the presence of Aldrithiol-2 and triphenyl phosphine in pyridine at ca. 70 °C, to afford the diester S37.6.
  • the diesters S37.3 are obtained by alkylation ofthe phosphonic acid S34.6 with a haloester S37.1.
  • the alkylation reaction is performed as described in Scheme 36 for the preparation ofthe esters S36.4. This method is illustrated in Scheme 37, Example 2.
  • the phosphonic acid S34.6 is reacted with excess ethyl 3-bromo-2- methylpropionate S37.7 and diisopropylethylamine in dimethylformamide at ca. 80 °C, as described in Anal Chem., 1987, 59, 1056, to produce the diester S37.8.
  • the diesters S37.3 are also obtained by displacement reactions of activated derivatives S34.7 ofthe phosphonic acid with the hydroxyesters S37.2.
  • the displacement reaction is performed in a polar solvent in the presence of a suitable base, as described in Scheme 36.
  • the displacement reaction is performed in the presence of an excess ofthe hydroxyester, to afford the diester product S37.3 in which the ester substituents are identical, or sequentially with limited amounts of different hydroxyesters, to prepare diesters S37.3 in which the ester substituents are different.
  • the methods are illustrated in Scheme 37, Examples 3 and 4. As shown in Example 3, the phosphoryl dichloride S35.22 is reacted with three molar equivalents of ethyl 3-hydroxy-2-(hydroxymethyl)propionate S37.9 in tetrahydrofuran containing potassium carbonate, to obtain the diester product S37.10.
  • 2,2-Dimethyl-2-aminoethylphosphonic acid intermediates can be prepared by the route in Scheme 38.
  • Condensation of 2-methyl-2- propanesulfina ide with acetone give sulfinyl imine S38.ll (J. Org. Chem. 1999, 64, 12).
  • Addition of dimethyl methylphosphonate lithium to S38.ll afford S38.12.
  • Acidic methanolysis of S38.12 provide amine S38.13. Protection of amine with Cbz group and removal of methyl groups yield phosphonic acid S38.14, which can be converted to desired S38.15 (Scheme 38a) using methods reported earlier on.
  • An alternative synthesis of compound S38.14 is also shown in Scheme 38b.
  • 2-Amino-6-chloropurine is alkylated at the N-9 position by heating with 3-(t- butyldimethylsilyloxy)phenethyl iodide and sodium hydride in DMF, following a procedure similar to that described in US patent application 2002/0068721.
  • the 2-amino group is converted to the iodo group by a conventional method such as that described in J. Med. Chem. 2003, 46, 5763.
  • the resulting iodide is cross-coupled with cyclopentylzinc bromide in the presence of a palladium catalyst such as bis(triphenylphosphine)palladium(II) chloride (J. Org. Chem. 1991, 56, 1445).
  • Transformation to the desired (4- ⁇ 2-cyclo ⁇ entyl-9-[2-(3- hydroxyphenyl)ethyl]-9H-purin-6-ylamino)phenoxymethyl)phosphonic acid diethyl ester is achieved by displacing the 6-chloro substituent with the corresponding phosphonate-containing aniline under reaction conditions such as those described in US patent application 2002/0068721, and then removing the t- butyldimethylsilyl protecting group by exposure to tetrabutylammonium fluoride.
  • Another specific compound ofthe invention can be synthesized as follows:
  • A-420983 is demethylated by condensing with ⁇ -chloroethyl chloroformate in the presence of Hunig's base in a solvent such as chloroform, followed by brief heating in acidic methanol.
  • the resulting free piperazine is alkylated with diethyl 2-bromoethylphosphonate in the presence of a base such as potassium carbonate, in a solvent such as dimethylformamide, to provide the desired product.
  • All literature and patent citations herein are hereby expressly incorporated by reference at the locations of their citation. Specifically cited sections or pages ofthe above cited works are incorporated by reference with specificity.

Abstract

The invention is related to phosphorus substituted kinase inhibitory compounds and conjugates, compositions containing such compounds and conjugates, and therapeutic methods that include the administration of such compounds and conjugates, as well as to processes and intermediates useful for preparing such compounds and conjugates.

Description

KINASE INHIBITOR PHOSPHONATE CONJUGATES
PRIORITY OF INVENTION This application claims the benefit of priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application Serial Number 60/622,962, filed 26 October 2004; and this application claims the benefit of priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application Serial Number 60/531932, filed 22 December 2003; and this application also claims priority to United States Patent Application Serial Number 10/832811 and to PCT Application Number PCT/US2004/013062, both filed 26 April 2004. The entirety of each ofthe above referenced applications is incorporated herein by reference. FIELD OF THE INVENTION The invention relates generally to phosphonate-containing compounds with kinase-inhibitory activity, i.e., compounds that inhibit at least one kinase. BACKGROUND OF THE INVENTION Improving the delivery of drugs and other agents to target cells and tissues has been the focus of considerable research for many years. Though many attempts have been made to develop effective methods for importing biologically active molecules into cells, both in vivo and in vitro, none has proved to be entirely satisfactory. Optimizing the association ofthe inhibitory drug with its intracellular target, while minimizing intercellular redistribution of the drug, e.g., to neighboring cells, is often difficult or inefficient. Most agents currently administered to a patient parenterally are not targeted, thereby resulting in systemic delivery ofthe agent to cells and tissues o he body where the agent is unnecessary, and often undesirable. This systemic delivery may result in adverse side effects and often limits the dose of an agent (e.g., glucocorticoids and other anti-inflammatory agents) that can be administered. By comparison, oral administration of agents is generally recognized as a convenient and economical method of administration. However, oral administration of agents can result in (a) the uptake ofthe agent through cellular and tissue barriers, such as the blood-brain barrier, epithelial, or the cell membrane, resulting in undesirable systemic distribution, and/or (b) temporary residence ofthe agent within the gastrointestinal tract. Accordingly, a major goal has been to develop methods for specifically targeting agents to cells and tissues. Benefits of such treatment includes avoiding the general physiological effects of inappropriate delivery of such agents to other cells and tissues, such as uninfected cells. Thus, there is a need for therapeutic agents, for example, agents that inhibit at least one kinase, with improved pharmacological properties, e.g., drugs having improved kinase-inhibitory activity and pharmacokinetic properties, including improved oral bioavailability, greater potency and extended effective half-life in vivo. Such inhibitors would have therapeutic uses, for example, as anti-cancer agents. Thus, new kinase inhibitors should have fewer side effects, less complicated dosing schedules, and be orally active. In particular, there is a need for a less onerous dosage regimen, such as one pill, once per day. Assay methods capable of determining the presence, absence or amounts of kinase inhibition are of practical utility in the search for kinase inhibitors as well as for diagnosing the presence of conditions associated with kinase activity. SUMMARY OF THE INVENTION Intracellular targeting may be achieved by methods and compositions that allow accumulation or retention of biologically active agents inside cells. The present invention provides novel analogs of kinase-inhibitory compounds, i.e., compounds that inhibit the activity of at least one kinase. Such novel kinase-inhibitory analogs possess utilities ofthe kinase-inhibitory compounds and optionally provide cellular accumulation. In addition, the present invention provides compositions and methods useful for inhibiting at least one kinase that may have therapeutic activity against diseases associated with kinase activity, such as cancer. The present invention relates generally to the accumulation or retention of therapeutic compounds inside cells. The invention is more particularly related to attaining high concentrations of phosphonate-containing molecules in target cells. Such effective targeting may be applicable to a variety of therapeutic formulations and procedures. Compounds ofthe invention include kinase-inhibitory compounds having at least one phosphonate group. Accordingly, in one embodiment the invention provides a conjugate comprising a kinase inhibiting compound linked to one or more phosphonate groups; or a pharmaceutically acceptable salt or solvate thereof. In another embodiment the invention provides a compound comprising one or more phosphonates and a substructure of formula I:
I wherein L1 and L2 are -N- or -CRa-; and Ra is hydrogen, alkyl, substituted alkyl, aryl or substituted aryl; or a pharmaceutically acceptable salt thereo. In another embodiment, the invention provides compound comprising one or more phosphonates and a substructure of formula II:
π
In another embodiment, the invention provides compound comprising one or more phosphonates and a substructure of formula Ilia, INa or Va: ma IVa Va
In another embodiment, the invention provides compound comprising one or more phosphonates and a substructure of formula III, IN or N:
In another embodiment, the invention provides a compound of any one of formulae 1-4:
wherein: A A0 is A1; A1 is
A3 is:
Y1 is independently O, S, N(RX), N(ORx), or N(N(RX)( Rx)); Y2 is independently a bond, O, N(RX), N(ORx), N(N(RX)( Rx)), or - S(O)M2~; and when Y2 joins two phosphorous atoms Y2 can also be C(R2)(R2);
Rx is independently H, R , W , a protecting group, or the formula:
Ry is independently H, W3, R2 or a protecting group; R2 is independently H, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R3 groups; R3 is R3a, R3b, R3° or R3d, provided that when R3 is bound to a heteroatom, then R3 is R3c or R3d; R3a is F, Cl, Br, I, -CN, N3 or -NO2; R3b is Y1; R3c is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), - S(O)2(ORx), -OC(Y1)Rx, -OC(Y1)ORx, -OC(Y1)(N(Rx)(Rx)), -SC(Y1)RX, - SC(Y*-)ORx, -SC(Y1)(N(RX)(RX)), -N(RX)C(Y1)RX, -N(Rx)C(Y1)ORx, or - N(RX)C(Y1)(N(RX)(RX)) ; R3d is -C(Y1)RX, -C(Y*-)ORx or -C(Y1)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; W3 is WVw5; W4 is R5, -CO^R5, -CtY^W5, -SO2R5, or -SO2W5; W5 is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R2 groups; W6 is W3 independently substituted with 1, 2, or 3 A3 groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mia, Ml c, and Mid are independently 0 or 1; M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; L1 and L2 are independently -N-, or -CRa-, provided that only one of L1 or L2 is a nitrogen atom; Ra is hydrogen, alkyl, aryl or substituted aryl; R20 is hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl aryl, cycloalkyl, substituted aryl, or -NR^0; Rb and Rc are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, or aralkyl; R21 is hydrogen, alkyl, cycloalkyl, substituted cycloalkyl, substituted alkyl, aryl, substituted aryl, aralkyl, or substituted aralkyl; and R22 and R23 are independently hydrogen, alkyl, substituted aryl, or aralkyl. In another embodiment, the invention provides a pharmaceutical composition comprising an effective amount of a compound ofthe invention and a pharmaceutically acceptable excipient. In another embodiment, the invention also provides a method of increasing cellular accumulation and retention of drug compounds, thus improving their therapeutic and diagnostic value, comprising linking the compound to one or more (e.g., 1, 2, 3, or 4) phosphonate groups. In another embodiment, the invention provides a method of inhibiting the activity of at least one kinase in an animal (e.g. a mammal) comprising administering an effective amount of a compound ofthe invention to the animal. In another embodiment, the invention provides a unit dosage form comprising a compound ofthe invention and a pharmaceutically acceptable excipient. In another embodiment, the invention provides a method for inhibiting a kinase in vitro or in vivo comprising contacting a sample in need of such treatment with a compound ofthe invention. In another embodiment, the invention provides a method of treating cancer in an animal (e.g. a mammal) in need of such treatment comprising administering an effective amount of a compound ofthe invention to the animal. In another embodiment, the invention provides a compound ofthe invention for use in medical therapy (preferably for use in treating a condition associated with kinase activity, e.g., elevated kinase activity), as well as the use of a compound of the invention for the manufacture of a medicament useful for the treatment of a condition associated with kinase activity, e.g., associated with elevated kinase activity. In another embodiment, the invention provides the use of a compound as described in any one of claims 1-55 to prepare a medicament for inhibiting a kinase in an animal (e.g. a mammal). In another embodiment, the invention provides the use of a compound of the invention to prepare a medicament for treating cancer in an animal (e.g. a mammal). In another embodiment, the invention provides a method for preparing a compound ofthe invention as described in the schemes and examples herein. In another embodiment, the invention provides a method for preparing a pharmaceutical composition, comprising combining a pharmaceutically acceptable excipient and a compound ofthe invention. In another embodiment, the invention provides processes and novel intermediates disclosed herein which are useful for preparing compounds of he invention. Some ofthe compounds ofthe invention are useful to prepare other compounds ofthe invention. In another aspect ofthe invention, the activity of a kinase is inhibited by a method comprising the step of treating a sample suspected of containing a kinase with a compound or composition of the invention. DETAILED DESCRIPTION OF THE INVNETION Reference will now be made in detail to certain embodiments ofthe invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope ofthe present invention as defined by the embodiments. Many ofthe current treatment regimes for cell proliferation diseases such as psoriasis and cancer utilize compounds that inhibit DNA synthesis. Such compounds are toxic to cells generally, but their toxic effect on rapidly dividing cells such as tumor cells can be beneficial. Alternative approaches to antiproliferative agents that act by mechanisms other than the inhibition of DNA synthesis have the potential to display enhanced selectivity of action. In recent years it has been discovered that a cell may become cancerous by virtue ofthe transformation of a portion of its DNA into an oncogene i.e. a gene that, on activation, leads to the formation of malignant tumor cells (Bradshaw, Mutagenesis 1986, 1, 91). Several such oncogenes give rise to the production of peptides which are receptors for growth factors. The growth factor receptor complex subsequently leads to an increase in cell proliferation. It is known, for example, that several oncogenes encode tyrosine kinase enzymes and that certain growth factor receptors are also tyrosine kinase enzymes (Yarden et al, Ann. Rev. Biochem., 1988, 57, 443; Larsen et al, Ann. Reports in Med. Chem. 1989, Chpt. 13). Receptor tyrosine kinases are important in the transmission of biochemical signals that initiate cell replication. They are large enzymes that span the cell membrane and possess an extracellular binding domain for growth factors such as epidermal growth factor (EGF), and an intracellular portion that functions as a kinase to phosphorylate tyrosine amino acids in proteins and hence influence cell proliferation. Various classes of receptor tyrosine kinases are known (Wilks, Advances in Cancer Research, 1993, 60, 43-73) based on families of growth factors that bind to different receptor tyrosine kinases. The classification includes Class I receptor tyrosine kinases comprising the EGF family of receptor tyrosine kinases such as the EGF, TGFα, NEU, erbB, Xmrk, HER and let23 receptors, Class II receptor tyrosine kinases comprising the insulin family of receptor tyrosine kinases such as the insulin, IGFI and insulin- related receptor (IRR) receptors and Class III receptor tyrosine kinases comprising the platelet-derived growth factor (PDGF) family of receptor tyrosine kinases such as the PDGFα, PDGFβ and colony-stimulating factor I (CSF1) receptors. Class I kinases, such as the EGF family of receptor tyrosine kinases, are frequently present in common human cancers such as breast cancer (Sainsbury et al, Brit. J. Cancer, 1988, 58, 458; Guerin et al, Oncogene Res., 1988, 3, 21 and Klijn et al, Breast Cancer Res. Treat, 1994, 29, 73), non-small cell lung cancers (NSCLCs) including adenocarcinomas (Cerny et al, Brit. J. Cancer, 1986, 54, 265; Reubi et al, Int. J. Cancer, 1990, 45, 269; and Rusch et al, Cancer Research, 1993, 53, 2379) and squamous cell cancer ofthe lung
(Hendler et al, Cancer Cells, 1989, 7, 347), bladder cancer (Neal et al, Lancet, 1985, 366), oesophageal cancer (Mukaida et al, Cancer, 1991, 68, 142), gastrointestinal cancer such as colon, rectal or stomach cancer (Bolen et al, Oncogene Res., 1987, 1, 149), cancer ofthe prostate (Visakorpi et al, Histochem. J., 1992, 24, 481), leukaemia (Konaka et al, Cell, 1984, 37, 1035) and ovarian, bronchial or pancreatic cancer (European Patent Specification No. 0400586). As further human tumor tissues are tested for the EGF family of receptor tyrosine kinases, it is expected that their widespread prevalence will be established in further cancers such as thyroid and uterine cancer. It is also known that EGF type tyrosine kinase activity is rarely detected in normal cells, whereas it is more frequently detected in malignant cells (Hunter, Cell, 1987, 50, 823). EGF receptors that possess tyrosine kinase activity are overexpressed in many human cancers such as brain, lung squamous cell, bladder, gastric, breast, head and neck, oesophageal, gynaecological and thyroid tumors (W. J. Gullick, Brit. Med. Bull, 1991, 47, 87). Accordingly, an inhibitor of receptor tyrosine kinases would be of value as a selective inhibitor ofthe growth of mammalian cancer cells (Yaish et al.
Science, 1988, 242, 933). Support for this view is provided by the demonstration that erbstatin, an EGF receptor tyrosine kinase inhibitor, specifically attenuates the growth in athymic nude mice of a transplanted human mammary carcinoma that expresses EGF receptor tyrosine kinase but is without effect on the growth of another carcinoma that does not express EGF receptor tyrosine kinase (Toi et al, Eur. J. Cancer Clin. Oncol, 1990, 26, 722.) Various derivatives of styrene also possess tyrosine kinase inhibitory properties (European Patent Application Nos. 0 211 363, 0 304 493 and 0 322 738) and may be used as anti-tumor agents. The in vivo inhibitory effect of two such styrene derivatives that are EGF receptor tyrosine kinase inhibitors has been demonstrated against the growth of human squamous cell carcinoma inoculated into nude mice (Yoneda et al, Cancer Research, 1991, 51, 4430). Various known tyrosine kinase inhibitors are disclosed in a more recent review by T. R. Burke Jr. (Drugs ofthe Future, 1992, 77, 119). Kinase inhibitors have valuable pharmacological properties and can be used, for example, as anti-tumor drugs and as drugs against atherosclerosis. The phosphorylation of proteins has long been known as an important step in the differentiation and proliferation of cells. Phosphorylation is catalyzed by protein kinases that are divided into serine/threonine kinases and tyrosine kinases. The serine/threonine kinases include protein kinase C and the tyrosine kinases include PDGF (platelet-derived growth factor)-receptor tyrosine kinase and Bcr- Abl kinase. Chronic myelogenous Leukemia (CML) is a hematological stem cell disorder associated with a specific chromosomal translocation known as the Philadelphia chromosome that is detected in 95% of patients with CML and 20% with acute lymphocytic leukemia (ALL). The molecular consequences ofthe translocation is the fusion ofthe abl protooncogene to the bcr gene resulting in the production of an activated from of Abl tyrosine protein kinase. The Bcr-Abl protein is capable of inducing leukemias in mice, thus implicating the protein as the cause of these diseases. Thus, kinase inhibitors inhibit cellular kinases that are involved in disease states, for example, Bcr-Abl. As the tyrosine kinase activity ofthe Bcr-Abl protein is essential to its transforming ability, an inhibitor would be useful therapy for these disorders. In addition, kinase inhibitors prevent the development of resistance (multi-drug resistance) in cancer treatment with other chemotherapeutic drugs or remove existing resistance to other chemotherapeutic drugs. Two processes, the de novo formation of vessels from differentiating endothelial cells or angioblasts in the developing embryo (vasculogenesis) and the growth of new capillary vessels from existing blood vessels (angiogenesis), are involved in the development ofthe vascular systems of animal organs and tissues. Transient phases of new vessel formation (neovascularization) also occur in the adult body, for example, during the menstrual cycle, pregnancy and wound healing. On the other hand, a number of diseases are known to be associated with deregulated angiogenesis, for example, retinopathies, psoriasis, hemangioblastoma, hemangioma, and neoplastic diseases (e.g., solid tumors). The complex processes of vasculogenesis and angiogenesis have been found to involve a whole range of molecules, especially angiogenic growth factors and their endothelial receptors, as well as cell adhesion molecules. Recent findings show that at the center ofthe network regulating the growth and differentiation ofthe vascular system and its components, both during embryonic development and normal growth and in a wide number of pathological anomalies and diseases, lies the angiogenic factor known as vascular endothelial growth factor (VEGF), along with its cellular receptors (see Breier, G., et al, Trends in Cell Biology 6, 454-6 (1996) and the references cited therein). VEGF is a dimeric, disulfide-linked 46-kDa glycoprotein and is related to platelet-derived growth factor (PDGF). It is produced by normal cell lines and tumor cell lines, is an endothelial cell-specific mitogen, shows angiogenic activity in in vivo test systems (e.g. rabbit cornea), is chemotactic for endothelial cells and monocytes, and induces plasminogen activators in endothelial cells, which are then involved in the proteolytic degradation of extracellular matrix during the formation of capillaries. A number of isoforms of VEGF show comparable biological activity, but differ in the type of cells that secrete them and in their heparin-binding capacity. In addition, there are other members ofthe VEGF family, such as placenta growth factor (PLGF) and VEGF-C. VEGF receptors are transmembranous receptor tyrosine kinases. They are characterized by an extracellular domain with seven immunoglobulin-like domains and an intracellular tyrosine kinase domain. Various types of VEGF receptor are known, e.g. VEGFR-1, VEGFR-2, and VEGFR-3. A large number of human tumors, especially gliomas and carcinomas, express high levels of VEGF and its receptors. This has led to the hypothesis that the VEGF released by tumor cells could stimulate the growth of blood capillaries and the proliferation of tumor endothelium in a paracrine manner and thus, through the improved blood supply, accelerate tumor growth. Increased VEGF expression could explain the occurrence of cerebral edema in patients with glioma. Direct evidence ofthe role of VEGF as a tumor angiogenesis factor in vivo has been obtained from studies in which VEGF expression or VEGF activity was inhibited. This was achieved with antibodies that inhibit VEGF activity, with dominant-negative VEGFR-2 mutants that inhibited signal transduction, or with the use of antisense- VEGF RNA techniques. All approaches led to a reduction in the growth of glioma cell lines or other tumor cell lines in vivo as a result of inhibited tumor angiogenesis. In addition, hypoxia, a large number of growth factors and cytokines, e.g.
Epidermal Growth Factor, Transforming Growth Factor a, Transforming Growth Factor A, Interleukin 1, and Interleukin 6, induce the expression of VEGF in cell experiments. Angiogenesis is regarded as a prerequisite for those tumors that grow beyond a maximum diameter of about 1-2 mm; up to this limit, oxygen and nutrients may be supplied to the tumor cells by diffusion. Every tumor, regardless of its origin and its cause, is thus thought to be dependent on angiogenesis for its growth after it has reached a certain size. Three principal mechanisms play important parts in the activity of angiogenesis inhibitors against tumors: 1) inhibition ofthe growth of vessels, especially capillaries, into avascular resting tumors, with the result that there is no net tumor growth owing to the balance that is achieved between apoptosis and proliferation; 2) prevention ofthe migration of tumor cells owing to the absence of bloodflow to and from tumors; and 3) inhibition of endothelial cell proliferation, thus avoiding the paracrine growth-stimulating effect exerted on the surrounding tissue by the endothelial cells that normally line the vessels. Inhibitors of cyclin-dependent kinases, e.g., Alvocidib (US Patent No. 4,900,727; also known as flavopiridol) have been identified as a potentially useful therapeutic agents for a variety of cancers, including gastrointestinal and colon tumors, leukemias and myelomas (see, for example, Intl. J. Oncol, 1996, 9, 1143). Inhibitors of tyrosine kinases, including Bcr-Abl, e.g., Gleevec, are useful for the treatment of chronic myeloid leukemia (CML), and potentially for treatment of other cancers that express these kinases, including acute lymphocytic leukemia (ALL) and certain solid tumors. Gleevec was approved for the treatment of inoperable and/or metastatic malignant gastrointestinal stromal tumors (GISTs). Inhibitors of Flt3 tyrosine kinase, e.g. , CEP-701 (US Patent No.
4,923,986) and Midostaurin (US Patent No. 5,093,330), have potential utility for the treatment of a variety of cancers (Cancer Res., 1999, 59, 10). Inhibitors of MAP Erk kinase, e.g., PD-184352 (U.S. Patent No. 6,251,943), have been identified as potentially useful therapeutic agents for a variety of oncological disorders, including colon, breast, pancreatic and non- small-cell lung cancers (see, for example, Proc. Am. Soc. Clin. Oncol, 2003, 22, abstract 816). Other kinase inhibitors, e.g., doramapimod (U.S. Patent No. 6,319,921), have been identified as potentially useful therapeutic agents for the treatment of inflammatory diseases such as rheumatoid arthritis, psoriasis and Crohn's disease. Other kinase inhibitors, e.g., BAY-43-9006 (U.S. Publication No.
2002/0165394) have been identified as potentially useful therapeutic agents for a variety of cancers including gastrointestinal and colon tumors, leukemia and carcinoma (Curr. Pharm. Design, 2002, 8, 2269). Cytokine receptors are critical for the development and homeostasis of immune cells. These receptors all require the cytoplasmic tyrosine kinase JAK3 for signaling (Changelian, P. S. et al, Science, 2003, 302, 875). CP-690,550 (WO 02,096,909) is an orally available Janus kinase (JAK)-3 inhibitor, for the potential treatment of transplant rejection and psoriasis. Thus, there is a need for therapeutic agents that are kinase inhibitors with improved pharmacological properties, e.g. , drugs having improved kinase- inhibitory activity and pharmacokinetic properties, including improved oral bioavailability, greater potency and extended effective half-life in vivo. Such inhibitors would have therapeutic potential as, e.g., anticancer agents. The kinase inhibitory compounds provided herein, which meet such needs, may be used to treat breast cancer, non-small cell lung cancers (NSCLCs), adenocarcinomas, squamous cell cancer ofthe lung, oesophageal cancer, gastrointestinal cancer, colon cancer, rectal cancer, stomach cancer, prostate cancer, leukaemia, ovarian cancer, bronchial cancer, pancreatic cancer, thyroid cancer, uterine cancer, brain cancer, lung squamous cell cancer, bladder cancer, gastric cancer, head and neck cancer, gynaecological and thyroid tumors, to prevent the development of resistance (multi-drug resistance) in cancer treatment with other chemotherapeutic drugs or remove existing resistance to other chemotherapeutic drugs, retinopathies, hemangioblastoma, hemangioma, and neoplastic diseases, gliomas, to inhibit tumor angiogenesis, myelomas, chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), inoperable and/or metastatic malignant gastrointestinal stromal tumors (GISTs), treatment of inflammatory diseases such as rheumatoid arthritis, Crohn's disease, treatment of cell proliferation diseases, and for the treatment of transplant rejection and psoriasis.
DEFINITIONS Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings: When tradenames are used herein, applicants intend to independently include the tradename product and the active pharmaceutical ingredient(s) ofthe tradename product. "Bioavailability" is the degree to which the pharmaceutically active agent becomes available to the target tissue after the agent's introduction into the body. Enhancement ofthe bioavailability of a pharmaceutically active agent can provide a more efficient and effective treatment for patients because, for a given dose, more ofthe pharmaceutically active agent will be available at the targeted tissue sites. The terms "phosphonate" and "phosphonate group" include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to a heteroatom , 3) single-bonded to a heteroatom, and 4) single-bonded to another heteroatom, wherein each heteroatom can be the same or different. The terms "phosphonate" and "phosphonate group" also include functional groups or moieties that comprise a phosphorous in the same oxidation state as the phosphorous described above, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having the characteristics described above. For example, the terms "phosphonate" and "phosphonate group" include phosphonic acid, phosphonic monoester, phosphonic diester, phosphonamidate, and phosphonthioate functional groups. In one specific embodiment ofthe invention, the terms "phosphonate" and "phosphonate group" include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double- bonded to an oxygen, 3) single-bonded to an oxygen, and 4) single-bonded to another oxygen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteristics. In another specific embodiment ofthe invention, the terms "phosphonate" and "phosphonate group" include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to an oxygen, 3) single-bonded to an oxygen or nitrogen, and 4) single-bonded to another oxygen or nitrogen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteristics. The term "prodrug" as used herein refers to any compound that when administered to a biological system generates the drug substance, i.e. active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, and/or metabolic chemical reaction(s). A prodrug is thus a covalently modified analog or latent form of a therapeutically- active compound. "Prodrug moiety" refers to a labile functional group that separates from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, "Design and Application of Prodrugs" in A Textbook of Drug Design and Development (1991), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academic Publishers, pp. 113-191). Enzymes that are capable of an enzymatic activation mechanism with the phosphonate prodrug compounds ofthe invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphases. Prodrug moieties can serve to enhance solubility, absorption and lipophilicity to optimize drug delivery, bioavailability and efficacy. A prodrug moiety may include an active metabolite or drug itself. Exemplary prodrug moieties include the hydrolytically sensitive or labile acyloxymethyl esters -CH2OC(=O)R9 and acyloxymethyl carbonates -CH2OC(=O)OR9 where R9 is d-C6 alkyl, C1-Q5 substituted alkyl, C6-C20 aryl or C6-C2o substituted aryl. The acyloxyalkyl ester was first used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al. (1983) J. Pharm. Sci. 72: 324; also U.S. Patent Nos. 4816570, 4968788, 5663159 and 5792756. Subsequently, the acyloxyalkyl ester was used to deliver phosphonic acids across cell membranes and to enhance oral bioavailability. A close variant ofthe acyloxyalkyl ester, the alkoxycarbonyloxyalkyl ester (carbonate), may also enhance oral bioavailability as a prodrug moiety in the compounds ofthe combinations ofthe invention. An exemplary acyloxymethyl ester is pivaloyloxymethoxy, (POM) -CH2OC(=O)C(CH3)3. An exemplary acyloxymethyl carbonate prodrug moiety is pivaloyloxymethylcarbonate (POC) -CH2OC(=O)OC(CH3)3. The phosphonate group may be a phosphonate prodrug moiety. The prodrug moiety may be sensitive to hydrolysis, such as, but not limited to a pivaloyloxymethyl carbonate (POC) or POM group. Alternatively, the prodrug moiety may be sensitive to enzymatic potentiated cleavage, such as a lactate ester or a phosphonamidate-ester group. Aryl esters of phosphorus groups, especially phenyl esters, are reported to enhance oral bioavailability (De Lombaert et al. (1994) J. Med. Chem. 37: 498). Phenyl esters containing a carboxylic ester ortho to the phosphate have also been described (Khamnei and Torrence, (1996) J. Med. Chem. 39:4109- 4115). Benzyl esters are reported to generate the parent phosphonic acid. In some cases, substituents at the ort/*o-or/>αrα-position may accelerate the hydrolysis. Benzyl analogs with an acylated phenol or an alkylated phenol may generate the phenolic compound through the action of enzymes, e.g., esterases, oxidases, etc., which in turn undergoes cleavage at the benzylic C-O bond to generate the phosphoric acid and the quinone methide intermediate. Examples of this class of prodrugs are described by Mitchell et al. (1992) J. Chem. Soc. Perkin Trans. II 2345; Glazier WO 91/19721. Still other benzylic prodrugs have been described containing a carboxylic ester-containing group attached to the benzylic methylene (Glazier WO 91/19721). Thio-containing prodrugs are reported to be useful for the intracellular delivery of phosphonate drugs. These proesters contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide.
Deesterification or reduction ofthe disulfide generates the free thio intermediate which subsequently breaks down to the phosphoric acid and episulfide (Puech et al (1993) Antiviral Res., 22: 155-174; Benzaria et al. (1996) J. Med. Chem. 39: 4958). Cyclic phosphonate esters have also been described as prodrugs of phosphorus-containing compounds (Erion et al, US Patent No. 6312662). "Protecting group" refers to a moiety of a compound that masks or alters the properties of a functional group or the properties ofthe compound as a whole. Chemical protecting groups and strategies for protection/deprotection are well known in the art. See e.g.. Protective Groups in Organic Chemistry. Theodora W. Greene, John Wiley & Sons, Inc., New York, 1991. Protecting groups are often utilized to mask the reactivity of certain functional groups, to assist in the efficiency of desired chemical reactions, e.g. , making and breaking chemical bonds in an ordered and planned fashion. Protection of functional groups of a compound alters other physical properties besides the reactivity of the protected functional group, such as the polarity, lipophilicity (hydrophobicity), and other properties which can be measured by common analytical tools. Chemically protected intermediates may themselves be biologically active or inactive. Protected compounds may also exhibit altered, and in some cases, optimized properties in vitro and in vivo, such as passage through cellular membranes and resistance to enzymatic degradation or sequestration. In this role, protected compounds with intended therapeutic effects may be referred to as prodrugs. Another function of a protecting group is to convert the parental drug into a prodrug, whereby the parental drug is released upon conversion of the prodrug in vivo. Because active prodrugs may be absorbed more effectively than the parental drug, prodrugs may possess greater potency in vivo than the parental drug. Protecting groups are removed either in vitro, in the instance of chemical intermediates, or in vivo, in the case of prodrugs. With chemical intermediates, it is not particularly important that the resulting products after deprotection, e.g., alcohols, be physiologically acceptable, although in general it is more desirable if the products are pharmacologically innocuous. Any reference to any ofthe compounds ofthe invention also includes a reference to a physiologically acceptable salt thereof. Examples of physiologically acceptable salts ofthe compounds ofthe invention include salts derived from an appropriate base, such as an alkali metal (for example, sodium), an alkaline earth (for example, magnesium), ammonium and NX4 + (wherein X is C1-C4 alkyl). Physiologically acceptable salts of a hydrogen atom or an amino group include salts of organic carboxylic acids such as acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, sulfuric, phosphoric and sulfamic acids. Physiologically acceptable salts of a compound of an hydroxy group include the anion of said compound in combination with a suitable cation such as Na+ and NX (wherein X is independently selected from H or a C1-C4 alkyl group). For therapeutic use, salts of active ingredients ofthe compounds ofthe invention will be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base. However, salts of acids or bases which are not physiologically acceptable may also find use, for example, in the preparation or purification of a physiologically acceptable compound. All salts, whether or not derived form a physiologically acceptable acid or base, are within the scope ofthe present invention. As used herein, the term "substructure" refers to a residue wherein any hydrogen atom(s) or replaceable group(s) has been or can be removed to provide an open valence for the substitution of a group including a phosphonate group, e.g., the substructure is a scaffold, to which a substituent -link-P(O)(OR1)2 is attached. The substructures can have additional groups attached. For a kinase inhibiting compound that comprises at least one phosphonate group and a substructure, it is understood that the compound includes the substructure as at least part ofthe overall structure ofthe compound. "Alkyl" is C1-C18 hydrocarbon containing normal, secondary, or tertiary carbon atoms. Examples are methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1- propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-proρyl (i-Pr, i-propyl, -CH(CH3)2), 1 -butyl (n-Bu, n-butyl, -CH2CH2CH2CH3), 2-methyl-l -propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, -CH(CH3)CH2CH3), 2-methyl-2- propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-methyl-2- butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2),
3-methyl-l-butyl (-CH2CH2CH(CH3)2), 2-methyl-l -butyl (- CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl
(-CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2- methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (- CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (-
CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl- 3-pentyl (-CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (-
C(CH3)2CH(CH3)2), 3,3-dimethyl-2-butyl (-CH(CH )C(CH3))3. "Alkenyl" is C2-C1 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp2 double bond. Examples include, but are not limited to, ethylene or vinyl (-CH=CH2), allyl (-CH2CH=CH2), cyclopentenyl (-C5H7), and 5-hexenyl (-CH2 CH2CH2CH CH"=CH2). "Alkynyl" is C2-C18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. Examples include, but are not limited to, acetylenic (-C≡CH) and propargyl (-CH2C≡CH). "Alkylene" refers to a saturated, branched or straight chain or cyclic hydrocarbon radical of 1-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Typical alkylene radicals include, but are not limited to, methylene (-CH2-) 1,2-ethyl (-CH2CH2-), 1,3-propyl (-CH2CH2CH2-), 1,4-butyl (-CH2CH2CH2CH2-), and the like. "Alkenylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. Typical alkenylene radicals include, but are not limited to, 1,2-ethylene (-CH=CH-). "Alkynylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. Typical alkynylene radicals include, but are not limited to, acetylene (-C≡C-), propargyl (-CH2C≡C-), and 4-pentynyl (-CH2CH2CH2C≡CH-). "Aryl" means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like. "Arylalkyl" refers to an acyclic alkyl radical in which one ofthe hydrogen atoms bonded to a carbon atom, typically a terminal or sp carbon atom, is replaced with an aryl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-l-yl, , naphthylmethyl, 2-naphthylethan-l- yl, naphthobenzyl, 2-naphthophenylethan-l-yl and the like. The arylalkyl group comprises 6 to 20 carbon atoms, e.g., the alkyl moiety, including alkanyl, alkenyl or alkynyl groups, ofthe arylalkyl group is 1 to 6 carbon atoms and the aryl moiety is 5 to 14 carbon atoms. "Substituted alkyl," "substituted aryl," "substituted arylalkyl,"
"substituted cycloalkyl," etc., mean alkyl, aryl, arylalkyl, and cycloalkyl respectively, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent. Typical substituents include, but are not limited to, -X, -R, -O", -OR, -SR, -S", -NR2, -NR3, =NR, -CX3, -CN, -OCN, -SCN, -N=C=O, -NCS, -NO, -NO2, =N2, -N3, NC(*=O)R, -C(=O)R,
C(=O)NRR -S(-=O)2O_, -S(=O)2OH, -S(=O)2R, -OS(=O)2OR, -S(=O)2NR, - S(=O)R, -OP(=O)O2RR -P(=O)O2RR -P(=O)(O )2, -P(=O)(OH)2, -C(*=O)R, -C(*=O)X, -C(S)R, -C(O)OR, -C(O)O", -C(S)OR, -C(O)SR, -C(S)SR, -C(O)NRR, -C(S)NRR, -C(NR)NRR, where each X is independently a halogen: F, Cl, Br, or I; and each R is independently -H, alkyl, aryl, heterocycle, protecting group or prodrug moiety. Alkylene, alkenylene, and alkynylene groups may also be similarly substituted. "Heterocycle" as used herein includes, by way of example and not limitation, those heterocycles described in Paquette, Leo A.; Principles of Modern Heterocyclic Chemistrv (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; The Chemistrv of Heterocyclic Compounds. A Series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; andJ. Am. Chem. Soc. (1960) 82:5566. In one specific embodiment ofthe invention "heterocycle" includes a "carbocycle" as defined herein, wherein one or more (e.g., 1, 2, 3, or 4) carbon atoms have been replaced with a heteroatom (e.g., O, N, or S). Examples of heterocycles include, by way of example and not limitation, pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H- 1,2,5- thiadiazinyl, 2H,6H-l,5,2-dithiazinyl, thienyl, thianthrenyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathinyl, 2H-ρyrrolyl, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H- indolyl, lH-indazoly, purinyl, 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, β- carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, isatinoyl, and bis-tetrahydrofuranyl:
By way of example and not limitation, carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5- pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2- pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3- pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl. By way of example and not limitation, nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline. Still more typically, nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1 -pyrrolyl, 1 -imidazolyl, 1- pyrazolyl, and 1-piperidinyl. "Carbocycle" refers to a saturated, unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle. Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyclo [5,6] or [6,6] system. Examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1- cyclopent-1-enyl, l-cycloρent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1- cyclohex-1-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, phenyl, spiryl and naphfhyl. The term "cycloalkyl" refers to a Ci-Cis hydrocarbon containing one or more rings. The term "chiral" refers to molecules which have the property of non- superimposability ofthe mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner. The term "stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement ofthe atoms or groups in space. "Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography. "Enantiomers" refer to two stereoisomers of a compound which are non- superimposable mirror images of one another. The term "treatment" or "treating," to the extent it relates to a disease or condition includes preventing the disease or condition from occurring, inhibiting the disease or condition, eliminating the disease or condition, and/or relieving one or more symptoms ofthe disease or condition. Stereochemical definitions and conventions used herein generally follow S. P. Parker. Ed.. McGraw-Hill Dictionary of Chemical Terms (19841 McGraw- Hill Book Company, New York; and Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L or R and S are used to denote the absolute configuration ofthe molecule about its chiral center(s). The prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity. Protecting Groups In the context ofthe present invention, protecting groups include prodrug moieties and chemical protecting groups. Protecting groups are available, commonly known and used, and are optionally used to prevent side reactions with the protected group during synthetic procedures, i.e. routes or methods to prepare the compounds ofthe invention. For the most part the decision as to which groups to protect, when to do so, and the nature ofthe chemical protecting group "PG" will be dependent upon the chemistry ofthe reaction to be protected against (e.g., acidic, basic, oxidative, reductive or other conditions) and the intended direction ofthe synthesis. The PG groups do not need to be, and generally are not, the same if the compound is substituted with multiple PG. In general, PG will be used to protect functional groups such as carboxyl, hydroxyl, thio, or amino groups and to thus prevent side reactions or to otherwise facilitate the synthetic efficiency. The order of deprotection to yield free, deprotected groups is dependent upon the intended direction ofthe synthesis and the reaction conditions to be encountered, and may occur in any order as determined by the artisan. Various functional groups ofthe compounds ofthe invention maybe protected. For example, protecting groups for -OH groups (whether hydroxyl, carboxylic acid, phosphonic acid, or other functions) include "ether- or ester- forming groups". Ether- or ester-forming groups are capable of functioning as chemical protecting groups in the synthetic schemes set forth herein. However, some hydroxyl and thio protecting groups are neither ether- nor ester-forming groups, as will be understood by those skilled in the art, and are included with amides, discussed below. A very large number of hydroxyl protecting groups and amide-forming groups and corresponding chemical cleavage reactions are described in Protective Groups in Organic Synthesis. Theodora W. Greene (John Wiley & Sons, Inc., New York, 1991, ISBN 0-471-62301-6) ("Greene"). See also Kocienski, Philip J.; Protecting Groups (Georg Thieme Verlag Stuttgart, New York, 1994), which is incorporated by reference in its entirety herein. In particular Chapter 1, Protecting Groups: An Overview, pages 1-20, Chapter 2, Hydroxyl Protecting Groups, pages 21-94, Chapter 3, Diol Protecting Groups, pages 95-117, Chapter 4, Carboxyl Protecting Groups, pages 118-154, Chapter 5, Carbonyl Protecting Groups, pages 155-184. For protecting groups for carboxylic acid, phosphonic acid, phosphonate, sulfonic acid and other protecting groups for acids see Greene as set forth below. Such groups include by way of example and not limitation, esters, amides, hydrazides, and the like. Ether- and Ester-forming protecting groups Ester-forming groups include: (1) phosphonate ester-forming groups, such as phosphonamidate esters, phosphorothioate esters, phosphonate esters, and phosphon-bis-amidates; (2) carboxyl ester-forming groups, and (3) sulphur ester-forming groups, such as sulphonate, sulfate, and sulfinate. The phosphonate moieties ofthe compounds ofthe invention may or may not be prodrug moieties, i.e. they may or may be susceptible to hydrolytic or enzymatic cleavage or modification. Certain phosphonate moieties are stable under most or nearly all metabolic conditions. For example, a dialkylphosphonate, where the alkyl groups are two or more carbons, may have appreciable stability in vivo due to a slow rate of hydrolysis. Within the context of phosphonate prodrug moieties, a large number of structurally-diverse prodrugs have been described for phosphonic acids (Freeman and Ross in Progress in Medicinal Chemistry 34: 112-147 (1997) and are included within the scope ofthe present invention. An exemplary phosphonate ester-forming group is the phenyl carbocycle in substructure A3 having the formula:
wherein Ri maybe H or C -Cn alkyl; ml is 1, 2, 3, 4, 5, 6, 7 or 8, and the phenyl carbocycle is substituted with 0 to 3 R2 groups. Where Yi is O, a lactate ester is formed, and where Yi is N(R ), N(OR2) or N(N(R2)2, a phosphonamidate ester results. In its ester-forming role, a protecting group typically is bound to any acidic group such as, by way of example and not limitation, a -CO2H or
-C(S)OH group, thereby resulting in -CO2Rx where Rx is defined herein. Also, Rx for example includes the enumerated ester groups of WO 95/07920. Examples of protecting groups include: C3-C12 heterocycle (described above) or aryl. These aromatic groups optionally are polycyclic or monocyclic. Examples include phenyl, spiryl, 2- and 3-pyrrolyl, 2- and 3-thienyl, 2- and 4-imidazolyl, 2-, 4- and 5-oxazolyl, 3- and 4-isoxazolyl, 2-, 4- and 5-thiazolyl, 3-, 4- and 5-isothiazolyl, 3- and 4- pyrazolyl, 1-, 2-, 3- and 4-pyridinyl, and 1-, 2-, 4- and 5-pyrimidinyl, C3-C12 heterocycle or aryl substituted with halo, R1, R^O-Ci-C^ alkylene, C1-C12 alkoxy, CN, NO2, OH, carboxy, carboxyester, thiol, thioester, C1-C12 haloalkyl (1-6 halogen atoms), C2-C12 alkenyl or C2-C12 alkynyl. Such groups include 2-, 3- and 4-alkoxyphenyl (C1-C12 alkyl), 2-, 3- and 4- methoxyphenyl, 2-, 3- and 4-ethoxyphenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5- diethoxyphenyl, 2- and 3-carboethoxy-4-hydroxyphenyl, 2- and 3-ethoxy-4- hydroxyphenyl, 2- and 3-ethoxy-5-hydroxyphenyl, 2- and 3-ethoxy-6- hydroxyphenyl, 2-, 3- and 4-O-acetylphenyl, 2-, 3- and 4-dimethylaminophenyl, 2-, 3- and 4-methylmercaptophenyl, 2-, 3- and 4-halophenyl (including 2-, 3- and 4-fluorophenyl and 2-, 3- and 4-chlorophenyl), 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5-dimethylphenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5-biscarboxyethylphenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5-dimethoxyρhenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- and 3,5-dihalophenyl (including 2,4-difluorophenyl and 3,5-difluorophenyl), 2-, 3- and 4-haloalkylphenyl (1 to 5 halogen atoms, C1-C12 alkyl including 4- trifluoromethylphenyl), 2-, 3- and 4-cyanophenyl, 2-, 3- and 4-nitroρhenyl, 2-, 3- and 4-haloalkylbenzyl (1 to 5 halogen atoms, C1-C12 alkyl including 4- trifluoromethylbenzyl and 2-, 3- and 4-trichloromethylphenyl and 2-, 3- and 4- trichloromethylphenyl), 4-N-methylpiperidinyl, 3-N-methylpiperidinyl, 1- efhylpiperazinyl, benzyl, alkylsalicylphenyl (C1-C4 alkyl, including 2-, 3- and 4- ethylsalicylphenyl), 2-,3- and 4-acetylphenyl, 1,8-dihydroxynaphthyl (-CioHg- OH) and aryloxy ethyl [C6-C9 aryl (including phenoxy ethyl)], 2,2'- dihydroxybiphenyl, 2-, 3- and 4-N,N~dialkylaminophenol, -C6H4CH2-N(CH3)2, trimethoxybenzyl, triethoxybenzyl, 2-alkyl pyridinyl (C1.4 alkyl);
-cH2-o-c(0)- ; /0 ;C4 C8
esters of 2-carboxyphenyl; and C1-C4 alkylene-C3-C6 aryl (including benzyl, - CH2-pyrrolyl, -CH2-thienyl, -CH2-imidazolyl, -CH2-oxazolyl, -CH2-isoxazolyl, -CH2-thiazolyl, -CH2-isothiazolyl, -CH2-pyrazolyl, -CH2-pyridinyl and -CH2- pyrimidinyl) substituted in the aryl moiety by 3 to 5 halogen atoms or 1 to 2 atoms or groups selected from halogen, C1-C12 alkoxy (including methoxy and ethoxy), cyano, nitro, OH, C1-C12 haloalkyl (1 to 6 halogen atoms; including - CH2CCI3), C1-C12 alkyl (including methyl and ethyl), C -Cι2 alkenyl or C2-Cι alkynyl; alkoxy ethyl [Ci-Cβ alkyl including -CH2-CH2-O-CH3 (methoxy ethyl)]; alkyl substituted by any ofthe groups set forth above for aryl, in particular OH or by 1 to 3 halo atoms (including -CH3j -CH(CH3)2, -C(CH3)3, - CH2CH3, -(CH2)2CH3, -(CH2)3CH3, -(CH2)4CH3, -(CH2)5CH3, -CH2CH2F, - CH2CH2C1, -CH2CF3, and -CH2CC13); propylmorpholino, 2,3-dihydro-6-hydroxyindene, sesamol, catechol monoester, ■ CH2-C(O)-N(R1)2, -CH2-S(O)(Rl), -CH2-S(O)2(R1), -CH2-CH(OC(O)CH2R1)- CH2(OC(O)CH2R1), cholesteryl, enolpyruvate (HOOC-C(=CH2)-), glycerol; a 5 or 6 carbon monosaccharide, disaccharide or oligosaccharide (3 to 9 monosaccharide residues); triglycerides such as α-D-β-diglycerides (wherein the fatty acids composing glyceride lipids generally are naturally occurring saturated or unsaturated C6-26? Cβ.is or CO- 10 fatty acids such as linoleic, lauric, myristic, palmitic, stearic, oleic, palmitoleic, linolenic and the like fatty acids) linked to acyl ofthe parental compounds herein through a glyceryl oxygen ofthe triglyceride; phospholipids linked to the carboxyl group through the phosphate ofthe phospholipid; phthalidyl (shown in Fig. 1 of Clayton et al, Antimicrob. Agents Chemo.
(191 A) 5(6):670-671; cyclic carbonates such as (5-Rd-2-oxo-l,3-dioxolen-4-yl) methyl esters (Sakamoto et al, Chem. Pharm. Bull. (1984) 32(6)2241-2248) where Rd is Rl, R4 or aryl; and / — \ -O^QOJN O
The hydroxyl groups ofthe compounds of this invention optionally are substituted with one of groups III, IV or V disclosed in WO 94/21604, or with isopropyl. Table A lists examples of protecting group ester moieties that for example can be bonded via oxygen to -C(O)O- and -P(O)(O-)2 groups. Several amidates also are shown, which are bound directly to -C(O)- or -P(O)2- Esters of structures 1-5, 8-10 and 16, 17, 19-22 are synthesized by reacting the compound herein having a free hydroxyl with the corresponding halide (chloride or acyl chloride and the like) and N ,N-dicyclohexyl-N-morpholine carboxamidine (or another base such as DBU, triethylamine, CSCO3, N,N-dimethylaniline and the like) in DMF (or other solvent such as acetonitrile or N-methylpyrrolidone). When the compound to be protected is a phosphonate, the esters of structures 5-7, 11, 12, 21, and 23-26 are synthesized by reaction ofthe alcohol or alkoxide salt (or the corresponding amines in the case of compounds such as 13, 14 and 15) with the monochlorophosphonate or dichlorophosphonate (or another activated phosphonate).
TABLE A
1. -CH2-C(O)-N(Rι)2 * 10. -CH2-O-C(O)-C(CH3)3
2. -CH2-S(O)(Rι) 11. -CH2-CCI3
3. -CH2-S(O)2(Rι) 12. -C6H5
4. -CH2-O-C(O)-CH2-C6H5 13. -NH-CH2-C(O)O-CH2CH3
5. 3-cholesteryl 14. -N(CH3)-CH2-C(O)O-CH2CH3
6. 3 -pyridyl 15. -NHRi
7. N-ethylmorpholino 16. -CH2-O-C(O)-Ci0Hi5
8. -CH2-O-C(O)-C6H5 17. -CH2-O-C(O)-CH(CH3)2
9. -CH2-O-C(O)-CH2CH3 18. -CH2-C#H(OC(O)CH2Rι)-CH2- -(OC(O)CH2Rι)*
# - chiral center is (R), (S) or racemate. Other esters that are suitable for use herein are described in EP 632048. Protecting groups also includes "double ester" forming profunctionalities
such as -CH2OC(O)OCH3, -CH2SCOCH3, -CH2OCON(CH3)2, or alkyl- or aryl-acyloxyalkyl groups ofthe structure -CH(R1 or W5)O((CO)R37) or -CH(R1 or W5)((CO)OR38) (linked to oxygen ofthe acidic group) wherein R37 and R38 are alkyl, aryl, or alkylaryl groups (see U.S. Patent No. 4968788). Frequently R37 and R38 are bulky groups such as branched alkyl, ortho- substituted aryl, meta-substituted aryl, or combinations thereof, including normal, secondary, iso- and tertiary alkyls of 1-6 carbon atoms. An example is the pivaloyloxymethyl group. These are of particular use with prodrugs for oral administration. Examples of such useful protecting groups are alkylacyloxymethyl esters and their derivatives, including -
CH(CH2CH2OCH3)OC(O)C(CH3)3, ; -
CH2OC(O)Cι0Hi5, -CH2OC(O)C(CH3)3, -CH(CH2OCH3)OC(O)C(CH3)3, - CH(CH(CH3)2)OC(O)C(CH3)3, -CH2OC(O)CH2CH(CH3)2, - CH2OC(O)C6Hn, -CH2OC(O)C6H55 -CH2OC(O)CioHi5, -
CH2OC(O)CH2CH3, -CH2OC(O)CH(CH3)2 , -CH2OC(O)C(CH3)3 and - CH2OC(O)CH2C6H5. In some embodiments the protected acidic group is an ester ofthe acidic group and is the residue of a hydroxyl-containing functionality. In other embodiments, an amino compound is used to protect the acid ftxnctionality. The residues of suitable hydroxyl or amino-containing functionalities are set forth above or are found in WO 95/07920. Of particular interest are the residues of amino acids, amino acid esters, polypeptides, or aryl alcohols. Typical amino acid, polypeptide and carboxyl-esterified amino acid residues are described on pages 11 -18 and related text of WO 95/07920 as groups LI or L2. WO
95/07920 expressly teaches the amidates of phosphonic acids, but it will be understood that such amidates are formed with any ofthe acid groups set forth herein and the amino acid residues set forth in WO 95/07920. Typical esters for protecting acidic functionalities are also described in WO 95/07920, again understanding that the same esters can be formed with the acidic groups herein as with the phosphonate ofthe '920 publication. Typical ester groups are defined at least on WO 95/07920 pages 89-93 (under R31 or R35)5 the table on page 105, and pages 21-23 (as R). Of particular interest are esters of unsubstituted aryl such as phenyl or arylalkyl such benzyl, or hydroxy-, halo-, alkoxy-, carboxy- and/or alkylestercarboxy-substituted aryl or alkylaryl, especially phenyl, ortho-ethoxyphenyl, or C1-C4 alkylestercarboxyphenyl (salicylate C1-C12 alkylesters). The protected acidic groups, particularly when using the esters or amides of WO 95/07920, are useful as prodrugs for oral administration. However, it is not essential that the acidic group be protected in order for the compounds of this invention to be effectively administered by the oral route. When the compounds ofthe invention having protected groups, in particular amino acid amidates or substituted and unsubstituted aryl esters are administered systemically or orally they are capable of hydrolytic cleavage in vivo to yield the free acid. One or more ofthe acidic hydroxyls are protected. If more than one acidic hydroxyl is protected then the same or a different protecting group is employed, e.g., the esters may be different or the same, or a mixed amidate and ester may be used. Typical hydroxy protecting groups described in Greene (pages 14-118) include substituted methyl and alkyl ethers, substituted benzyl ethers, silyl ethers, esters including sulfonic acid esters, and carbonates. For example:
• Ethers (methyl, t-butyl, allyl);
• Substituted Methyl Ethers (Methoxymethyl, Methylthiomethyl, t- Butylthiomethyl, (Phenyldimethylsilyl)methoxymethyl, Benzyloxymethyl, p- Methoxybenzyloxymethyl, (4-Methoxyphenoxy)methyl, Guaiacolmethyl, t- Butoxymethyl, 4-Pentenyloxymethyl, Siloxymethyl, 2- Methoxyethoxymethyl, 2,2,2-Trichloroethoxymethyl, Bis(2- chloroethoxy)methyl, 2-(Trimethylsilyl)ethoxymefhyl, Tetrahydropyranyl, 3- Bromotetrahydropyranyl, Tetrahydropthiopyranyl, 1-Methoxycyclohexyl, 4- Methoxytetrahydropyranyl, 4-Methoxytetrahydrothiopyranyl, 4- Methoxytetrahydropthiopyranyl S,S-Dioxido, l-[(2-Chloro-4- methyl)ρhenyl]-4-methoxypiperidin-4-yl, 1 ,4-Dioxan-2-yl, Tetrahydrofuranyl, Tetrahydrothiofuranyl, 2,3,3a,4,5,6,7,7a-Octahydro- 7,8,8-trimethyl-4,7-methanobenzofuran-2-yl));
• Substituted Ethyl Ethers (1-Ethoxyethyl, 1 -(2-Chloroethoxy)ethyl, 1 -Methyl- 1-methoxyethyl, 1 -Methyl- 1-benzyloxyethyl, 1 -Methyl- l-benzyloxy-2- fluoroethyl, 2,2,2-Trichloroethyl, 2-Trimethylsilylethyl, 2- (Phenylselenyl)ethyl,
• ?-Chlorophenyl,^?-Methoxyphenyl, 2,4-Dinitrophenyl, Benzyl);
• Substituted Benzyl Ethers (p-Methoxybenzyl, 3,4-Dimethoxybenzyl, o- Nitrobenzyl, --Nitrobenzyl, 7-Halobenzyl, 2,6-Dichlorobenzyl, p- Cyanobenzyl, -Phenylbenzyl, 2- and 4-Picolyl, 3-Methyl-2-picolyl N- Oxido, Diphenylmethyl, * jp'-Dinitrobenzhydryl, 5-Dibenzosuberyl, Triphenylmethyl, α-Νaphthyldiphenylmethyl, p- methoxyphenyldiphenylmethyl, Di(p-methoxyphenyl)phenylmethyl, Tri( »- methoxyphenyl)methyl, 4-(4'-Bromophenacyloxy)phenyldiphenylmethyl, 4,4',4"-Tris(4,5-dichlorophthalimidophenyl)methyl, 4,4',4"- Tris(levulinoyloxyphenyl)methyl, 4,4',4"-Tris(benzoyloxyphenyl)methyl, 3- (hnidazol- 1 -ylmethyl)bis(4',4"-dimethoxyphenyl)methyl, 1 , 1 -Bis(4- methoxyphenyl)-l'-ρyrenylmethyl, 9-Anthryl, 9-(9-Phenyl)xanthenyl, 9-(9- Phenyl-10-oxo)anthryl, l,3-Benzodithiolan-2-yl, Benzisothiazolyl S,S- Dioxido); • Silyl Ethers (Trimethylsilyl, Triethylsilyl, Triisopropylsilyl, Dimethylisopropylsilyl, Diethylisopropylsilyl, Dimethylthexylsilyl, t- Butyldimethylsilyl, t-Butyldiphenylsilyl, Tribenzylsilyl, Tri-/?-xylylsilyl, Triphenylsilyl, Diphenylmethylsilyl, t-Butylmethoxyphenylsilyl);
• Esters (Formate, Benzoylformate, Acetate, Choroacetate, Dichloroacetate, Trichloroacetate, Trifluoroacetate, Methoxyacetate, Triphenylmethoxyacetate, Phenoxyacetate, --Chlorophenoxyacetate, p-poly- Phenylacetate, 3-Phenylpropionate, 4-Oxopentanoate (Levulinate), 4,4- (Ethylenedithio)pentanoate, Pivaloate, Adamantoate, Crotonate, 4- Methoxycrotonate, Benzoate, -Phenylbenzoate, 2,4,6-Trimethylbenzoate (Mesitoate));
• Carbonates (Methyl, 9-Fluorenylmethyl, Ethyl, 2,2,2-Trichloroethyl, 2- (Trimethylsilyl)ethyl, 2-(Phenylsulfonyl)ethyl, 2- (Triphenylphosphonio)ethyl, Isobutyl, Vinyl, Allyl, /-Nitrophenyl, Benzyl, -Methoxybenzyl, 3,4-Dimethoxybenzyl, o-Nifrobenzyl, j Nitrobenzyl, S- Benzyl Thiocarbonate, 4-Ethoxy-l-naphthyl, Methyl Dithiocarbonate);
• Groups With Assisted Cleavage (2-Iodobenzoate, 4-Azidobutyrate, 4-Nitro- 4-methylpentanoate, o-(Dibromomethyl)benzoate, 2- Formylbenzenesulfonate, 2-(Methylthiomethoxy)ethyl Carbonate, 4- (Methylthiomethoxy)butyrate, 2-(Methylthiomethoxymethyl)benzoate); Miscellaneous Esters (2,6-Dichloro-4-methylphenoxyacetate, 2,6-Dichloro- 4-(l,l,3,3 tetramethylbutyl)phenoxyacetate, 2,4-Bis(l,l- dimefhylρropyl)phenoxyacetate, Chlorodiphenylacetate, Isobutyrate, Monosuccinate, (E)-2-Methyl-2-butenoate (Tigloate), o- (Methoxycarbonyl)benzoate, jp-poly-Benzoate, α-Naphthoate, Nitrate, Alkyl NNN'N-Tetramethylphosphorodiamidate, N-Phenylcarbamate, Borate, Dimethylphosphinothioyl, 2,4-Dinitrophenylsulfenate); and • Sulfonates (Sulfate, Mefhanesulfonate (Mesylate), Benzylsulfonate, Tosylate). Typical 1,2-diol protecting groups (thus, generally where two OH groups are taken together with the protecting functionality) are described in Greene at pages 118-142 and include Cyclic Acetals and Ketals (Methylene, Ethylidene, 1- t-Butylethylidene, 1-Phenylethylidene, (4-Methoxyphenyl)ethylidene, 2,2,2- Trichloroethylidene, Acetonide (Isopropylidene), Cyclopentylidene, Cyclohexylidene, Cycloheptylidene, Benzylidene,/*-Methoxybenzylidene, 2,4- Dimethoxybenzylidene, 3,4-Dimethoxybenzylidene, 2-Νitrobenzylidene); Cyclic Ortho Esters (Methoxymethylene, Ethoxymethylene, Dimethoxymethylene, 1- Methoxyethylidene, 1-Ethoxyethylidine, 1,2-Dimethoxyethylidene, α-
Methoxybenzylidene, l-(NN-Dimethylamino)ethylidene Derivative, α -(NN- Dimethylamino)benzylidene Derivative, 2-Oxacyclopentylidene); Silyl Derivatives (Di-t-butylsilylene Group, 1,3-(1, 1,3,3-
Tetraisopropyldisiloxanylidene), and Tetra-t-butoxydisiloxane-l,3-diylidene), Cyclic Carbonates, Cyclic Boronates, Ethyl Boronate and Phenyl Boronate. More typically, 1,2-diol protecting groups include those shown in Table B, still more typically, epoxides, acetonides, cyclic ketals and aryl acetals.
Table B
wherein R^ is C1-C6 alkyl.
Amino protecting groups Another set of protecting groups include any ofthe typical amino protecting groups described by Greene at pages 315-385. They include:
• Carbamates: (methyl and ethyl, 9-fluorenylmethyl, 9(2- sulfo)fluorenylmethyl, 9-(2,7-dibromo)fluorenylmethyl, 2,7-di-t-butyl-[9- (10, 10-dioxo- 10, 10, 10, 10-tetrahydrothioxanthyl)]methyl, 4- methoxyphenacyl);
• Substituted Ethyl: (2,2,2-trichoroethyl, 2-trimethylsilylethyl, 2-phenylethyl, 1 -(1 -adamantyl)- 1 -methylethyl, 1 , 1 -dimethyl-2-haloethyl, 1 , 1 -dimethyl-2,2- dibromoethyl, l,l-dimethyl-2,2,2-trichloroethyl, 1 -methyl- 1 -(4- biρhenylyl)ethyl, 1 -(3, 5-di-t-butylphenyl)-l -methylethyl, 2-(2'- and 4*- pyridyl)ethyl, 2-(NN-dicyclohexylcarboxamido)ethyl, t-butyl, 1-adamantyl, vinyl, allyl, 1-isopropylallyl, cinnamyl, 4-nitrocinnamyl, 8-quinolyl, N- hydroxypiperidinyl, alkyldithio, benzyl, j?-methoxybenzyl,^-nitrobenzyl, 7- bromobenzyl, -chlorobenzyl, 2,4-dichlorobenzyl, 4-methylsulfinylbenzyl, 9-anthrylmethyl, diphenylmethyl);
• Groups With Assisted Cleavage: (2-methylthioethyl, 2-methylsulfonylethyl, 2-(p-toluenesulfonyl)ethyl, [2-(l ,3-dithianyl)]methyl, 4-methylthiophenyl, 2,4-dimethylthiophenyl, 2-phosphonioethyl, 2- triphenylphosphonioisopropyl, l,l-dimethyl-2-cyanoethyl, m-choro- - acyloxybenzyl, ?-(dihydroxyboryl)benzyl, 5-benzisoxazolylmethyl, 2- (trifluoromethyl)-6-chromonylmethyl);
• Groups Capable of Photolytic Cleavage: (m-nitrophenyl, 3,5- dimethoxybenzyl, o-nitrobenzyl, 3,4-dimethoxy-6-nitrobenzyl, phenyl(o- nitrophenyl)mefhyl); Urea-Type Derivatives (phenothiazinyl-(lθ)-carbonyl, N- ?-toluenesulfonylaminocarbonyl, N'-phenylaminothiocarbonyl);
• Miscellaneous Carbamates: (t-amyl, S-benzyl thiocarbamate,/"-cyanobenzyl, cyclobutyl, cyclohexyl, cyclopentyl, cyclopropylmethyl, -decyloxybenzyl, diisopropylmethyl, 2,2-dimethoxycarbonylvinyl, o-(N,N- dimethylcarboxamido)benzyl, 1 , 1 -dimethyl-3-(N,N- dimethylcarboxamido)propyl, 1,1-dimethylpropynyl, di(2-pyridyl)methyl, 2- furanylmethyl, 2-Iodoethyl, Isobornyl, Isobutyl, Isonicotinyl, j9-(f*'- Methoxyphenylazo)benzyl, 1-methylcyclobutyl, 1-methylcyclohexyl, 1- methyl- 1-cycloρroρylmethyl, 1 -methyl- 1 -(3, 5-dimethoxyphenyl)ethyl, 1- methyl- 1 -(p-phenylazophenyl)ethyl, 1 -methyl- 1 -phenyl ethyl, 1 -methyl- 1 -(4- pyridyl)efhyl, phenyl, j5-(phenylazo)benzyl, 2,4,6-tri-t-butylphenyl, 4- (trimetl ylammonium)benzyl, 2,4,6-trimethylbenzyl);
• Amides: (N-formyl, N-acetyl, N-choroacetyl, N-trichoroacetyl, N- trifluoroacetyl, N-phenylacetyl, N-3 -phenylpropionyl, N-picolinoyl, N-3 - pyridylcarboxamide, N-benzoylphenylalanyl, N-benzoyl, N-p- phenylbenzoyl);
• Amides With Assisted Cleavage: (N-o-nitrophenylacetyl, N-o- nitrophenoxyacetyl, N-acetoacetyl, (N - dithiobenzyloxycarbonylamino)acetyl, N-3 -(p-hydroxyphenyl)propionyl, N- 3-(o-nitrophenyl)propionyl, N-2-methyl-2-(o-nitrophenoxy)propionyl, N-2- mefhyl-2-(o-phenylazophenoxy)propionyl, N-4-chlorobutyryl, N-3-methyl-3- nitrobutyryl, N-o-nitrocinnamoyl, N-acetylmethionine, N-o-nitrobenzoyl, N- ø-(benzoyloxymethyl)benzoyl, 4,5-diphenyl-3 -oxazolin-2-one);
• Cyclic Imide Derivatives: (N-phthalimide, N-dithiasuccinoyl, N-2,3- diphenylmaleoyl, N-2,5-dimethylpyrrolyl, N- 1,1, 4,4- tetramethyldisilylazacyclopentane adduct, 5-substituted l,3-dimethyl-l,3,5- triazacyclohexan-2-one, 5-substituted 1 ,3-dibenzyl-l ,3-5-triazacyclohexan-2- one, 1-substituted 3,5-dinitro-4-pyridonyl);
• N- Alkyl and N-Aryl Amines: (N-methyl, N-allyl, N-[2- (trimethylsilyl)ethoxy]methyl, N-3-acetoxyproρyl, N-(l-isopropyl-4-nitro-2- oxo-3 -pyrrolin-3-yl), Quaternary Ammonium Salts, N-benzyl, N-di(4- methoxyphenyl)methyl, N-5-dibenzosuberyl, N-triphenylmethyl, N-(4- methoxyphenyl)diphenylmethyl, N-9-phenylfluorenyl, N-2,7-dichloro-9- fluorenylmethylene, N-ferrocenylmethyl, N-2-picolylamine N-oxide);
• hnine Derivatives: (N-l,l-dimethylthiomethylene, N-benzylidene, N-p- methoxybenylidene, N-diphenylmethylene, N- [(2-pyridyl)mesityl]methylene, N,(N,N-dimemylaminomethylene, NN-isopropylidene, N-p- nitrobenzylidene, N-salicylidene, N-5-chlorosalicylidene, N-(5-chloro-2- hydroxyphenyl)phenylmethylene, N-cyclohexylidene);
• Enamine Derivatives: (N-(5,5-dimethyl-3-oxo-l-cyclohexenyl)); • N-Metal Derivatives (N-borane derivatives, N-diphenylborinic acid derivatives, N-[phenyl(pentacarbonylchromium- or -tungsten)] carbenyl, N- copper orN-zinc chelate);
• Ν-Ν Derivatives: (N-nitro, N-nitroso, N-oxide);
• Ν-P Derivatives: (N-diphenylphosphinyl, N-dimethylthiophosphinyl, N- diphenylthiophosphinyl, N-dialkyl phosphoryl, N-dibenzyl phosphoryl, N- diphenyl phosphoryl);
• Ν-Si Derivatives, Ν-S Derivatives, and Ν-Sulfenyl Derivatives: (N- benzenesulfenyl, N-o-nitrobenzenesulfenyl, N-2,4-dinitrobenzenesulfenyl, N- pentachlorobenzenesulfenyl, N-2-nitro-4-methoxybenzenesulfenyl, N- triphenylmethylsulfenyl, N-3-nitropyridinesulfenyl); and N-sulfonyl Derivatives (N-p-toluenesulfonyl, N-benzenesulfonyl, N-2,3,6-trimethyl-4- methoxybenzenesulfonyl, N-2,4,6-trimethoxybenzenesulfonyl, N-2,6- dimethyl-4-methoxybenzenesulfonyl, N-pentamethylbenzenesulfonyl, N- 2,3,5,6,-tetramethyl-4-methoxybenzenesulfonyl, N-4- methoxybenzenesulfonyl, N-2,4,6-trimethylbenzenesulfonyl, N-2,6- dimethoxy-4-methylbenzenesulfonyl, N-2,2,5,7,8-pentamethylchroman-6- sulfonyl, N-methanesulfonyl, N-β-trimethylsilyethanesulfonyl, N-9- anthracenesulfonyl, N-4-(4',8'-dimethoxynaphthylmethyl)benzenesulfonyl, N-benzylsulfonyl, N-trifluoromethylsulfonyl, N-phenacylsulfonyl). More typically, protected amino groups include carbamates and amides, still more typically, -ΝHC(O)R1 or -N=CR1N(R1)2- Another protecting group, also useful as a prodrug for amino or -NH(R5), is:
See for example Alexander, J. et al. (1996) J. Med. Chem. 39:480-486.
Amino acid and polypeptide protecting group and conjugates An amino acid or polypeptide protecting group of a compound ofthe invention has the structure R15NHCH(R16)C(O)-, where R15 is H, an amino acid or polypeptide residue, or R5, and R16 is defined below. R16 is lower alkyl or lower alkyl (Ci-Cβ) substituted with amino, carboxyl, amide, carboxyl ester, hydroxyl, C6-C7 aryl, guanidinyl, imidazolyl, indolyl, sulfhydryl, sulfoxide, and/or alkylphosphate. R10 also is taken together with the amino acid α N to form a proline residue (R10 = -(CH2)3-). However, R10 is generally the side group of a naturally-occurring amino acid such as H, - CH3, -CH(CH3)2, -CH2-CH(CH3)25 -CHCH3-CH2-CH3, -CH2-C6H5, -CH2CH2- S-CH3, -CH2OH, -CH(OH)-CH3, -CH2-SH, -CH2-C6H4OH, -CH2-CO-NH2, - CH2-CH2-CO-NH2, -CH2-COOH, -CH2-CH2-COOH, -(CH2)4-NH2 and - (CH2)3-NH-C(NH2)-NH2. Rio also includes l-guanidinoprop-3-yl, benzyl, 4- hydroxybenzyl, imidazol-4-yl, indol-3-yl, methoxyphenyl and ethoxyphenyl. Another set of protecting groups include the residue of an amino- containing compound, in particular an amino acid, a polypeptide, a protecting group, -NΗSO2R NHC(O)R, -N(R)2, NH2 or -NH(R)(H), whereby for example a carboxylic acid is reacted, i.e. coupled, with the amine to form an amide, as in C(O)NR2. A phosphonic acid may be reacted with the amine to form a phosphonamidate, as in -P(O)(OR)(NR2). In general, amino acids have the structure R17C(O)CH(R16)NH-, where
R is -OH, -OR, an amino acid or a polypeptide residue. Amino acids are low molecular weight compounds, on the order of less than about 1000 MW and which contain at least one amino or imino group and at least one carboxyl group. Generally the amino acids will be found in nature, i.e., can be detected in biological material such as bacteria or other microbes, plants, animals or man. Suitable amino acids typically are alpha amino acids, i.e. compounds characterized by one amino or imino nitrogen atom separated from the carbon atom of one carboxyl group by a single substituted or unsubstituted alpha carbon atom. Of particular interest are hydrophobic residues such as mono-or di-alkyl or aryl amino acids, cycloalkylamino acids and the like. These residues contribute to cell permeability by increasing the partition coefficient ofthe parental drug. Typically, the residue does not contain a sulfhydryl or guanidino substituent. Naturally-occurring amino acid residues are those residues found naturally in plants, animals or microbes, especially proteins thereof. Polypeptides most typically will be substantially composed of such naturally- occurring amino acid residues. These amino acids are glycine, alanine, valine, leucine, isoleucine, serine, threonine, cysteine, methionine, glutamic acid, aspartic acid, Iysine, hydroxylysine, arginine, histidine, phenylalanine, tyrosine, tryptophan, proline, asparagine, glutamine and hydroxyproline. Additionally, unnatural amino acids, for example, valanine, phenylglycine and homoarginine are also included. Commonly encountered amino acids that are not gene- encoded may also be used in the present invention. All ofthe amino acids used in the present invention may be either the D- or L- optical isomer. In addition, other peptidomimetics are also useful in the present invention. For a general review, see Spatola, A. F., in Chemistrv and Biochemistry of Amino Acids. Peptides and Proteins, B. Weinstein, eds., Marcel Dekker, New York, p. 267 (1983). When protecting groups are single amino acid residues or polypeptides they optionally are substituted at R of substituents A , A or A in a compound ofthe invention. These conjugates are produced by forming an amide bond between a carboxyl group ofthe amino acid (or C-terminal amino acid of a polypeptide for example). Similarly, conjugates are formed between R3 and an amino group of an amino acid or polypeptide. Generally, only one of any site in the parental molecule is amidated with an amino acid as described herein, although it is within the scope of this invention to introduce amino acids at more than one permitted site. Usually, a carboxyl group of R3 is amidated with an amino acid. In general, the α-amino or α-carboxyl group ofthe amino acid or the terminal amino or carboxyl group of a polypeptide are bonded to the parental functionalities, i.e., carboxyl or amino groups in the amino acid side chains generally are not used to form the amide bonds with the parental compound (although these groups may need to be protected during synthesis ofthe conjugates as described further below). With respect to the carboxyl-containing side chains of amino acids or polypeptides it will be understood that the carboxyl group optionally will be blocked, e.g., by R1, esterified with R5 or amidated. Similarly, the amino side chains R16 optionally will be blocked with R1 or substituted with R5. Such ester or amide bonds with side chain amino or carboxyl groups, like the esters or amides with the parental molecule, optionally are hydrolyzable in vivo or in vitro under acidic (pH <3) or basic (pH >10) conditions.
Alternatively, they are substantially stable in the gastrointestinal tract of humans but are hydrolyzed enzymatically in blood or in intracellular environments. The esters or amino acid or polypeptide amidates also are useful as intermediates for the preparation ofthe parental molecule containing free amino or carboxyl groups. The free acid or base ofthe parental compound, for example, is readily formed from the esters or amino acid or polypeptide conjugates of this invention by conventional hydrolysis procedures. When an amino acid residue contains one or more chiral centers, any of the D, L, meso, threo or erythro (as appropriate) racemates, scalemates or mixtures thereof may be used. In general, if the intermediates are to be hydrolyzed non-enzymatically (as would be the case where the amides are used as chemical intermediates for the free acids or free amines), D isomers are useful. On the other hand, L isomers are more versatile since they can be susceptible to both non-enzymatic and enzymatic hydrolysis, and are more efficiently transported by amino acid or dipeptidyl transport systems in the gastrointestinal tract. Examples of suitable amino acids whose residues are represented by Rx or Ry include the following: Glycine; Aminopolycarboxylic acids, e.g., aspartic acid, β-hydroxyaspartic acid, glutamic acid, β -hydroxyglutamic acid, β-methylaspartic acid, β-methylglutamic acid, β, β-dimethylaspartic acid, γ-hydroxyglutamic acid, β, γ-dihydroxyglutamic acid, β -phenylglutamic acid, γ-methyleneglutamic acid, 3-aminoadipic acid, 2- aminopimelic acid, 2-aminosuberic acid and 2-aminosebacic acid; Amino acid amides such as glutamine and asparagine; Polyamino- or polybasic-monocarboxylic acids such as arginine, Iysine, β -aminoalanine, γ -aminobutyrine, ornithine, citruline, homoarginine, homocitrulline, hydroxylysine, allohydroxylsine and diaminobutyric acid; Other basic amino acid residues such as histidine; Diaminodicarboxylic acids such as α, α'-diaminosuccinic acid, α, α'- diaminoglutaric acid, α, α'-diaminoadipic acid, α, α'-diaminopimelic acid, α, α'- diamino- β-hydroxypimelic acid, α, α'-diaminosuberic acid, α, α'-diaminoazelaic acid, and α, α'-diaminosebacic acid; Imino acids such as proline, hydroxyproline, allohydroxyproline, γ- methylproline, pipecolic acid, 5-hydroxypipecolic acid, and azetidine-2- carboxylic acid; A mono- or di-alkyl (typically Ci-Cs branched or normal) amino acid such as alanine, valine, leucine, allylglycine, butyrine, norvaline, norleucine, heptyline, α-methylserine, α-amino-α-methyl-γ-hydroxyvaleric acid, α-amino- α- methyl-δ-hydroxyvaleric acid, α-amino- α-methyl-ε-hydroxycaproic acid, isovaline, α-methylglutamic acid, α-aminoisobutyric acid, α-aminodiethylacetic acid, α-aminodiisopropylacetic acid, α-aminodi-n-propylacetic acid, α- aminodiisobutylacetic acid, α-aminodi-n-butylacetic acid, α- aminoethylisopropylacetic acid, α-amino-n-propylacetic acid, α- aminodiisoamyacetic acid, α-methylaspartic acid, α-methylglutamic acid, 1- aminocyclopropane-1 -carboxylic acid, isoleucine, alloisoleucine, tert-leucine, β- methyltryptophan and α-amino- β-ethyl-β-phenylpropionic acid; β-phenylserinyl; Aliphatic α-amino-β-hydroxy acids such as serine, β-hydroxyleucine, β- hydroxynorleucine, β -hydroxynorvaline, and α-amino-β-hydroxystearic acid; α- Amino, α-, γ-, δ- or ε-hydroxy acids such as homoserine, δ - hydroxynorvaline, γ-hydroxynorvaline and ε-hydroxynorleucine residues; canavine and canaline; γ -hydroxyornithine; 2-hexosaminic acids such as D-glucosaminic acid or D-galactosaminic acid; α-Amino-β-thiols such as penicillamine, β-thiolnorvaline or β- thiolbutyrine; Other sulfur containing amino acid residues including cysteine; homocystine, β-phenylmethionine, methionine, S-allyl-L-cysteine sulfoxide, 2- thiolhistidine, cystathionine, and thiol ethers of cysteine or homocysteine; Phenylalanine, tryptophan and ring-substituted α-amino acids such as the phenyl- or cyclohexylamino acids α-aminophenylacetic acid, α- aminocyclohexylacetic acid and α-amino-β-cyclohexylpropionic acid; phenylalanine analogues and derivatives comprising aryl, lower alkyl, hydroxy, guanidino, oxyalkylether, nitro, sulfur or halo-substituted phenyl (e.g., tyrosine, methyltyrosine and o-chloro-, p-chloro-, 3,4-dichloro, o-, m- or/?-methyl-, 2,4,6- trimethyl-, 2-ethoxy-5-nitro-, 2-hydroxy-5-nitro- and p-nitro-phenylalanine); furyl-, thienyl-, pyridyl-, pyrimidinyl-, purinyl- or naphthyl-alanines; and tryptophan analogues and derivatives including kynurenine, 3- hydroxykynurenine, 2-hydroxytryptophan and 4-carboxytryptophan; α-Amino substituted amino acids including sarcosine (N-methylglycine), N-benzylglycine, N-mefhylalanine, N-benzylalanine, N-methylphenylalanine, N- benzylphenylalanine, N-methylvaline and N-benzylvaline; and α-Hydroxy and substituted α -hydroxy amino acids including serine, threonine, allothreonine, phosphoserine and phosphothreonine. Polypeptides are polymers of amino acids in which a carboxyl group of one amino acid monomer is bonded to an amino or imino group ofthe next amino acid monomer by an amide bond. Polypeptides include dipeptides, low molecular weight polypeptides (about 1500-5000 MW) and proteins. Proteins optionally contain 3, 5, 10, 50, 75, 100 or more residues, and suitably are substantially sequence-homologous with human, animal, plant or microbial proteins. They include enzymes (e.g., hydrogen peroxidase) as well as immunogens such as KLH, or antibodies or proteins of any type against which one wishes to raise an immune response. The nature and identity ofthe polypeptide may vary widely. The polypeptide amidates are useful as immunogens in raising antibodies against either the polypeptide (if it is not immunogenic in the animal to which it is administered) or against the epitopes on the remainder ofthe compound of this invention. Antibodies capable of binding to the parental non-peptidyl compound are used to separate the parental compound from mixtures, for example in diagnosis or manufacturing ofthe parental compound. The conjugates of parental compound and polypeptide generally are more immunogenic than the polypeptides in closely homologous animals, and therefore make the polypeptide more immunogenic for facilitating raising antibodies against it. Accordingly, the polypeptide or protein may not need to be immunogenic in an animal typically used to raise antibodies, e.g., rabbit, mouse, horse, or rat, but the final product conjugate should be immunogenic in at least one of such animals. The polypeptide optionally contains a peptidolytic enzyme cleavage site at the peptide bond between the first and second residues adjacent to the acidic heteroatom. Such cleavage sites are flanked by enzymatic recognition structures, e.g., a particular sequence of residues recognized by a peptidolytic enzyme. Peptidolytic enzymes for cleaving the polypeptide conjugates of this invention are well known, and in particular include carboxypeptidases. Carboxypeptidases digest polypeptides by removing C-terminal residues, and are specific in many instances for particular C-terminal sequences. Such enzymes and their substrate requirements in general are well known. For example, a dipeptide (having a given pair of residues and a free carboxyl terminus) is covalently bonded through its α-amino group to the phosphorus or carbon atoms ofthe compounds herein. In embodiments where Wi is phosphonate it is expected that this peptide will be cleaved by the appropriate peptidolytic enzyme, leaving the carboxyl ofthe proximal amino acid residue to autocatalytically cleave the phosphonoamidate bond. Suitable dipeptidyl groups (designated by their single letter code) are AA, AR, AN, AD, AC, AE, AQ, AG, AH, AI, AL, AK, AM, AF, AP, AS, AT, AW, AY, AV, RA, RR, RN, RD, RC, RE, RQ, RG, RH, Rl, RL, RK, RM, RF, RP, RS, RT, RW, RY, RV, NA, NR, NN, ND, NC, NE, NQ, NG, NH, NI, NL, NK, NM, NF, NP, NS, NT, NW, NY, NV, DA, DR, DN, DD, DC, DE, DQ, DG, DH, DI, DL, DK, DM, DF, DP, DS, DT, DW, DY, DV, CA, CR, CN, CD, CC, CE, CQ, CG, CH, CI, CL, CK, CM, CF, CP, CS, CT, CW, CY, CV, EA, ER, EN, ED, EC, EE, EQ, EG, EH, El, EL, EK, EM, EF, EP, ES, ET, EW, EY, EV, QA, QR, QN, QD, QC, QE, QQ, QG, QH, Ql, QL, QK, QM, QF, QP, QS, QT, QW, QY, QV, GA, GR, GN, GD, GC, GE, GQ, GG, GH, GI, GL, GK, GM, GF, GP, GS, GT, GW, GY, GV, HA, HR, HN, HD, HC, HE, HQ, HG, HH, HI, HL, HK, HM, HF, HP, HS, HT, HW, HY, HV, IA, IR, IN, ID, IC, IE, IQ, IG, IH, II, IL, IK, IM, IF, IP, IS, IT, IW, IY, IV, LA, LR, LN, LD, LC, LE, LQ, LG, LH, LI, LL, LK, LM, LF, LP, LS, LT, LW, LY, LV, KA, KR, KN, KD, KC, KE, KQ, KG, KH, KI, KL, KK, KM, KF, KP, KS, KT, KW, KY, KV, MA, MR, MN, MD, MC, ME, MQ, MG, MH, MI, ML, MK, MM, MF, MP, MS, MT, MW, MY, MV, FA, FR, FN, FD, FC, FE, FQ, FG, FH, FI, FL, FK, FM, FF, FP, FS, FT, FW, FY, FV, PA, PR, PN, PD, PC, PE, PQ, PG, PH, PI, PL, PK, PM, PF, PP, PS, PT, PW, PY, PV, SA, SR, SN, SD, SC, SE, SQ, SG, SH, SI, SL, SK, SM, SF, SP, SS, ST, SW, SY, SV, TA, TR, TN, TD, TC, TE, TQ, TG, TH, TI, TL, TK, TM, TF, TP, TS, TT, TW, TY, TV, WA, WR, WN, WD, WC, WE, WQ, WG, WH, WI, WL, WK, WM, WF, WP, WS, WT, WW, WY, WV, YA, YR, YN, YD, YC, YE, YQ, YG, YH, YI, YL, YK, YM, YF, YP, YS, YT, YW, YY, YV, VA, VR, VN, VD, VC, VE, VQ, VG, VH, VI, VL, VK, VM, VF, VP, VS, VT, VW, VY and W. Tripeptide residues are also useful as protecting groups. When a phosphonate is to be protected, the sequence -X4-pro-X5- (where X4 is any amino acid residue and X5 is an amino acid residue, a carboxyl ester of proline, or hydrogen) will be cleaved by luminal carboxypeptidase to yield X4 with a free carboxyl, which in turn is expected to autocatalytically cleave the phosphonoamidate bond. The carboxy group of X5 optionally is esterified with benzyl. Dipeptide or tripeptide species can be selected on the basis of known transport properties and/or susceptibility to peptidases that can affect transport to intestinal mucosal or other cell types. Dipeptides and tripeptides lacking an α- amino group are transport substrates for the peptide transporter found in brush border membrane of intestinal mucosal cells (Bai, J.P.F., (1992) Pharm Res. 9:969-978). Transport competent peptides can thus be used to enhance bioavailability ofthe amidate compounds. Di- or tripeptides having one or more amino acids in the D configuration are also compatible with peptide transport and can be utilized in the amidate compounds of this invention. Amino acids in the D configuration can be used to reduce the susceptibility of a di- or tripeptide to hydrolysis by proteases common to the brush border such as aminopeptidase N. In addition, di- or tripeptides alternatively are selected on the basis of their relative resistance to hydrolysis by proteases found in the lumen ofthe intestine. For example, tripeptides or polypeptides lacking asp and/or glu are poor substrates for aminopeptidase A, di- or tripeptides lacking amino acid residues on the N-terminal side of hydrophobic amino acids (leu, tyr, phe, val, trp) are poor substrates for endopeptidase, and peptides lacking a pro residue at the penultimate position at a free carboxyl terminus are poor substrates for carboxypeptidase P. Similar considerations can also be applied to the selection of peptides that are either relatively resistant or relatively susceptible to hydrolysis by cytosolic, renal, hepatic, serum or other peptidases. Such poorly cleaved polypeptide amidates are immunogens or are useful for bonding to proteins in order to prepare immunogens. Specific Embodiments ofthe Invention Specific values described for radicals, substituents, and ranges, as well as specific embodiments ofthe invention described herein, are for illustration only; they do not exclude other defined values or other values within defined ranges. In one specific embodiment ofthe invention A1 is ofthe formula:
In another specific embodiment ofthe invention A1 is ofthe formula:
In another specific embodiment ofthe invention A1 is ofthe formula:
In another specific embodiment ofthe invention A1 is ofthe formula:
In another specific embodiment ofthe invention A1 is ofthe formula:
and W τ-5a is a carbocycle or a heterocycle where W 5aa . is independently substituted with 0 or 1 R groups. A specific value for Ml 2a is 1. In another specific embodiment ofthe invention A1 is ofthe formula:
In another specific embodiment ofthe invention A1 is ofthe formula:
In another specific embodiment ofthe invention A is ofthe formula:
wherein W5a is a carbocycle independently substituted with 0 or 1 R2 groups; In another specific embodiment ofthe invention A1 is ofthe formula:
wherein Y2b is O or N(R2); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention A1 is ofthe formula:
wherein W5a is a carbocycle independently substituted with 0 or 1 R2 groups; In another specific embodiment ofthe invention A1 is ofthe formula:
wherein W5a is a carbocycle or heterocycle where W5a is independently substituted with 0 or 1 R2 groups. In another specific embodiment ofthe invention A1 is ofthe formula:
wherein Y2b is O or N(R2); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In a specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Yla is O or S; and Y2a is O, N(RX) or S. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y rZbD . is O or N(RX). In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y τ-2bD is O or N(RX); and M12d is 1, 2, 3, 4, 5, , 7 or 8. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y2b is O or N(RX); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention Ml 2d is 1. In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention W5 is a carbocycle. In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention W5 is phenyl. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Yla is O or S; and Y2a is O, N(RX) or S. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y2 is O or N(RX). hi another specific embodiment ofthe invention A is ofthe formula:
wherein Y2b is O or N(RX); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention R1 is H. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein the phenyl carbocycle is substituted with 0, 1, 2, or 3 R2 groups. In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Yla is O or S; and Y2a is O, N(R2) or S. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Yla is O or S; Y2b is O or N(R2); and Y2c is O, N(Ry) or S. In another specific embodiment ofthe invention A3 is ofthe formula:
wheren Yla is O or S; Y2b is O or N(R2); Y2d is O or N(Ry); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y2b is O or N(R2); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y2b is O or N(R2). In another specific embodiment ofthe invention A3 is ofthe formula:
In another specific embodiment ofthe invention A is ofthe formula:
In another specific embodiment ofthe invention A is ofthe formula:
wherein Yla is O or S; and Y2a is O, N(R2) or S. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Yla is O or S; Y2b is O or N(R2); and Y2c is O, N(Ry) or S.
In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Yla is O or S; Y2b is O or N(R2); Y2d is O or N(Ry); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y2 is O or N(R2); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein Y2 is O or N(R2).
In another specific embodiment ofthe invention A is ofthe formula:
wherein: Y2b is O or N(RX); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein the phenyl carbocycle is substituted with 0, 1, 2, or 3 R groups. In another specific embodiment ofthe invention A3 is ofthe formula:
wherein the phenyl carbocycle is substituted with 0, 1, 2, or 3 R 2 groups In another specific embodiment ofthe invention A is ofthe formula:
In a specific embodiment ofthe invention A0 is ofthe formula:
wherein each R is independently (C1-C6)alkyl. In a specific embodiment ofthe invention Rx is independently H, R , W , a protecting group, or the formula:
wherein: Ry is independently H, W3, R2 or a protecting group; R1 is independently H or alkyl of 1 to 18 carbon atoms; R2 is independently H, R1, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R3 groups or taken together at a carbon atom, two R2 groups form a ring of 3 to 8 carbons and the ring maybe substituted with 0 to 3 R3 groups; In a specific embodiment ofthe invention Rx is ofthe formula:
wherein Yla is O or S; and Y2c is O, N(Ry) or S. In a specific embodiment ofthe invention Rx is ofthe formula:
wherein Yla is O or S; and Y2d is O or N(Ry). In a specific embodiment ofthe invention Rx is ofthe formula:
In a specific embodiment of the invention Ry is hydrogen or alkyl of 1 to
10 carbons. In a specific embodiment ofthe invention Rx is ofthe formula:
In a specific embodiment ofthe invention Rx is ofthe formula:
In a specific embodiment ofthe invention Rx is ofthe formula:
In a specific embodiment ofthe invention Y1 is O or S In a specific embodiment ofthe invention Y2 is O, N(Ry) or S. In one specific embodiment ofthe invention Rx is a group ofthe formula:
wherein: mla, mlb, mlc, mid and mle are independently 0 or 1; ml2c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Ry is H, W3, R2 or a protecting group; provided that: if mla, ml2c, and mid are 0, then mlb, mlc and mle are 0; if mla and ml2c are 0 and mid is not 0, then mlb and mlc are 0; if mla and mid are 0 and ml2c is not 0, then mlb and at least one of mlc and mle are 0; if mla is 0 and ml2c and mid are not 0, then mlb is 0; if ml2c and mid are 0 and mla is not 0, then at least two of mlb, mlc and mle are 0; if ml2c is 0 and mla and mid are not 0, then at least one of mlb and mlc are 0; and if mid is 0 and mla and ml2c are not 0, then at least one of mlc and mle are 0. In compounds ofthe invention W5 carbocycles and W heterocycles may be independently substituted with 0 to 3 R2 groups. W5 may be a saturated, unsaturated or aromatic ring comprising a mono- or bicyclic carbocycle or heterocycle. W5 may have 3 to 10 ring atoms, e.g., 3 to 7 ring atoms. The W5 rings are saturated when containing 3 ring atoms, saturated or mono-unsaturated when containing 4 ring atoms, saturated, or mono- or di-unsaturated when containing 5 ring atoms, and saturated, mono- or di-unsaturated, or aromatic when containing 6 ring atoms. A W5 heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S). W5 heterocyclic monocycles may have 3 to 6 ring atoms (2 to 5 carbon atoms and 1 to 2 heteroatoms selected from N, O, and S); or 5 or 6 ring atoms (3 to 5 carbon atoms and 1 to 2 heteroatoms selected from N and S). W5 heterocyclic bicycles have 7 to 10 ring atoms (6 to 9 carbon atoms and 1 to 2 heteroatoms selected from N, O, and S) arranged as a bicyclo [4,5], [5,5], [5,6], or [6,6] system; or 9 to 10 ring atoms (8 to 9 carbon atoms and 1 to 2 hetero atoms selected from N and S) arranged as a bicyclo [5,6] or [6,6] system. The W5 heterocycle may be bonded to Y2 through a carbon, nitrogen, sulfur or other atom by a stable covalent bond. W5 heterocycles include for example, pyridyl, dihydropyridyl isomers, piperidine, pyridazinyl, pyrimidinyl, pyrazinyl, s-triazinyl, oxazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, furanyl, thiofuranyl, thienyl, and pyrrolyl. W5 also includes, but is not limited to, examples such as:
W5 carbocycles and heterocycles may be independently substituted with 0 to 3 R2 groups, as defined above. For example, substituted W5 carbocycles include:
Examples of substituted phenyl carbocycles include:
Linking Groups and Linkers The invention provides conjugates that comprise a kinase inhibiting compound that is linked to one or more phosphonate groups either directly (e.g. through a covalent bond) or through a linking group (i.e. a linker). The nature of the linker is not critical provided it does not interfere with the ability ofthe phosphonate containing compound to function as a therapeutic agent. The phosphonate or the linker can be linked to the compound (e.g. a compound of 100-103) at any synthetically feasible position on the compound by removing a hydrogen or any portion ofthe compound to provide an open valence for attachment ofthe phosphonate or the linker. In one embodiment of the invention the linking group or linker (which π 1 can be designated "L") can include all or a portions ofthe group A , A , A , or W3 described herein. In another embodiment ofthe invention the linking group or linker has a molecular weight of from about 20 daltons to about 400 daltons. In another embodiment ofthe invention the linking group or linker has a length of about 5 angstroms to about 300 angstroms. In another embodiment ofthe invention the linking group or linker separates the DRUG and a P(=Y1) residue by about 5 angstroms to about 200 angstroms, inclusive, in length. In another embodiment ofthe invention the linking group or linker is a divalent, branched or unbranched, saturated or unsaturated, hydrocarbon chain, having from 2 to 25 carbon atoms, wherein one or more (e.g. 1, 2, 3, or 4) ofthe carbon atoms is optionally replaced by (-O-), and wherein the chain is optionally substituted on carbon with one or more (e.g. 1, 2, 3, or 4) substituents selected from (Ci-C6)alkoxy, (C3-C6)cycloalkyl, (C1-C6)alkanoyl, ( -C^alkanoyloxy, ( -C^alkoxycarbonyl, ( -C^alkylthio, azido, cyano, nitro, halo, hydroxy, oxo (=O), carboxy, aryl, aryloxy, heteroaryl, and heteroaryloxy. In another embodiment ofthe invention the linking group or linker is of the formula W-A wherein A is ( -C^alkyl, (C2-C24)alkenyl, (C2-C24)alkynyl, (C3-C8)cycloalkyl, (Ce-C^aryl or a combination thereof, wherein W is - N(R)C(=O)-, -C(=O)N(R)-, -OC(=O)-, -C(=O)O-, -O-, -S-, -S(O)-, -S(O)2-, - N(R)-, -C(*=O)-, or a direct bond; wherein each R is independently H or (Ci- C6)alkyl. In another embodiment ofthe invention the linking group or linker is a divalent radical formed from a peptide. In another embodiment ofthe invention the linking group or linker is a divalent radical formed from an amino acid. In another embodiment ofthe invention the linking group or linker is a divalent radical formed from poly-L-glutamic acid, poly-L-aspartic acid, poly-L- histidine, poly-L-ornithine, poly-L-serine, poly-L-threonine, poly-L-tyrosine, poly-L-leucine, poly-L-lysine-L-phenylalanine, poly-L-lysine or poly-L-lysine- L-tyrosine. In another embodiment ofthe invention the linking group or linker is of the formula W-(CH2)n wherein, n is between about 1 and about 10; and W is - N(R)C(*=O)-, -C(=O)N(R)-, -OC(=O)-, -C(=O)O-, -O-, -S-, -S(O)-, -S(O)2-, - C(*=O)-, -N(R)-, or a direct bond; wherein each R is independently H or ( - C6)alkyl. In another embodiment ofthe invention the linking group or linker is methylene, ethylene, or propylene. In another embodiment ofthe invention the linking group or linker is attached to the phosphonate group through a carbon atom ofthe linker. Intracellular Targeting The phosphonate group ofthe compounds ofthe invention may cleave in vivo in stages after they have reached the desired site of action, i.e. inside a cell. One mechanism of action inside a cell may entail a first cleavage, e.g. by esterase, to provide a negatively-charged "locked-in" intermediate. Cleavage of a terminal ester grouping in a compound ofthe invention thus affords an unstable intermediate which releases a negatively charged "locked in" intermediate. After passage inside a cell, intracellular enzymatic cleavage or modification ofthe phosphonate or prodrug compound may result in an intracellular accumulation ofthe cleaved or modified compound by a "trapping" mechanism. The cleaved or modified compound may then be "locked-in" the cell by a significant change in charge, polarity, or other physical property change which decreases the rate at which the cleaved or modified compound can exit the cell, relative to the rate at which it entered as the phosphonate prodrug. Other mechanisms by which a therapeutic effect are achieved may be operative as well. Enzymes which are capable of an enzymatic activation mechanism with the phosphonate prodrug compounds ofthe invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphatases. From the foregoing, it will be apparent that many different drugs can be derivatized in accord with the present invention. Numerous such drugs are specifically mentioned herein. However, it should be understood that the discussion of drug families and their specific members for derivatization according to this invention is not intended to be exhaustive, but merely illustrative. Kinase-inhibitory Compounds The compounds ofthe invention include those with kinase-inhibitory activity. The compounds ofthe inventions bear one or more (e.g. 1, 2, 3, or 4) phosphonate groups, which may be a prodrug moiety. The term "kinase-inhibitory compound" includes those compounds that inhibit the activity of at least one kinase. In particular, the compounds include CP-690,550, AP23464, A-420983 and roscovitine. Typically, compounds ofthe invention have a molecular weight of from about 400 amu to about 10,000 amu; in a specific embodiment ofthe invention, compounds have a molecular weight of less than about 5000 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 2500 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 1000 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 800 amu; in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 600 amu; and in another specific embodiment ofthe invention, compounds have a molecular weight of less than about 600 amu and a molecular weight of greater than about 400 amu. The compounds ofthe invention also typically have a logD (polarity) less than about 5. In one embodiment the invention provides compounds having a logD less than about 4; in another embodiment the invention provides compounds having a logD less than about 3; in another embodiment the invention provides compounds having a logD greater than about -5; in another embodiment the invention provides compounds having a logD greater than about -3; and in another embodiment the invention provides compounds having a logD greater than about 0 and less than about 3. Selected substituents within the compounds ofthe invention are present to a recursive degree. In this context, "recursive substituent" means that a substituent may recite another instance of itself. Because ofthe recursive nature of such substituents, theoretically, a large number may be present in any given embodiment. For example, Rx contains a Ry substituent. Ry can be R2, which in turn can be R3. If R3 is selected to be R3°, then a second instance of Rx can be selected. One of ordinary skill in the art of medicinal chemistry understands that the total number of such substituents is reasonably limited by the desired properties ofthe compound intended. Such properties include, by way of example and not limitation, physical properties such as molecular weight, solubility or log P, application properties such as activity against the intended target, and practical properties such as ease of synthesis. By way of example and not limitation, W3, Ry and R3 are all recursive substituents in certain embodiments. Typically, each of these may independently occur 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or 0, times in a given embodiment. More typically, each of these may independently occur 12 or fewer times in a given embodiment. More typically yet, W3 will occur 0 to 8 times, Ry will occur 0 to 6 times and R3 will occur 0 to 10 times in a given embodiment. Even more typically, W3 will occur 0 to 6 times, Ry will occur 0 to 4 times and R3 will occur 0 to 8 times in a given embodiment. Recursive substituents are an intended aspect ofthe invention. One of ordinary skill in the art of medicinal chemistry understands the versatility of such substituents. To the degree that recursive substituents are present in an embodiment ofthe invention, the total number will be determined as set forth above. Whenever a compound described herein is substituted with more than one ofthe same designated group, e.g., "R1" or "R6a"5 then it will be understood that the groups may be the same or different, i.e., each group is independently selected. Wavy lines indicate the site of covalent bond attachments to the adjoining groups, moieties, or atoms. In one embodiment ofthe invention, the compound is in an isolated and purified form. Generally, the term "isolated and purified" means that the compound is substantially free from biological materials (e.g. blood, tissue, cells, etc.). In one specific embodiment ofthe invention, the term means that the compound or conjugate ofthe invention is at least about 50 wt.% free from biological materials; in another specific embodiment, the term means that the compound or conjugate ofthe invention is at least about 75 wt.% free from biological materials; in another specific embodiment, the term means that the compound or conjugate ofthe invention is at least about 90 wt.% free from biological materials; in another specific embodiment, the term means that the compound or conjugate ofthe invention is at least about 98 wt.% free from biological materials; and in another embodiment, the term means that the compound or conjugate ofthe invention is at least about 99 wt.% free from biological materials. In another specific embodiment, the invention provides a compound or conjugate ofthe invention that has been synthetically prepared (e.g., ex vivo). In one embodiment ofthe invention, the compound is not an anti- inflammatory compound; in another embodiment the compound is not an anti- infective; in another embodiment the compound is not a compound that is active against immune-mediated conditions; in another embodiment the compound is not a compound that is active against metabolic diseases; in another embodiment the compound is not an antiviral agent; in another embodiment the compound is not a nucleoside; in another embodiment the compound is not a IMPDH inhibitor; in another embodiment the compound is not an antimetabolite; in another embodiment the compound is not a PNP inhibitor; in another embodiment the compound inhibits a serine/threonine kinase, tyrosine kinase, Bcr-Abl kinase, cyclin-dependent kinase, Flt3 tyrosine kinase, MAP Erk kinase, JAK3 kinase, VEGF receptor kinase, PDGF receptor tyrosine kinase, protein kinase C, insulin receptor tyrosine kinase, or an EGF receptor tyrosine kinase; in another embodiment the compound is not Gefitinib, imatinib, erlotinib, vatalanib, alvocidib, CEP-701, GLEEVEC, midostaurin, MLN-518, PD-184352, doramapimod, BAY-43-9006, or CP-690,550; in another embodiment the compound is not a compound of any one of formulae 1-4. In one embodiment the invention provides a conjugate comprising a kinase inhibiting compound linked to one or more phosphonate groups; or a pharmaceutically acceptable salt or solvate thereof, wherein the kinase inhibiting compound is not Gefitinib, imatinib, erlotinib, vatalanib, alvocidib, CEP-701, GLEEVEC, midostaurin, MLN-518, PD-184352, doramapimod, BAY-43-9006, or CP-690,550. In another embodiment, the invention provides a compound of any one of formulae 500-511:
511
that is substituted with one or more groups A0, wherein: A is A , A2 or W3 with the proviso that the conjugate includes at least one A A1 is:
A2 is:
A3 is: Y1 is independently O, S, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), or N(N(RX)( RX)); Y2 is independently a bond, O, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), N(N(RX)( Rx)), -S(O)M2-, or -S(O)M2-S(O)M2-; and when Y2 joins two 9 9 9 phosphorous atoms Y can also be C(R )(R ); Rx is independently H, R1, R2, W3, a protecting group, or the formula:
wherein: Ry is independently H, W3, R2 or a protecting group; R1 is independently H or alkyl of 1 to 18 carbon atoms; R2 is independently H, R1, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R groups or taken together at a carbon atom, two R groups form a ring of 3 to 8 carbons and the ring may be substituted with 0 to 3 R3 groups; R3 is R3a, R3b, R3c or R3d, provided that when R3 is bound to a heteroatom, then R3 is R3c or R3d; R3a is F, Cl, Br, I, -CN, N3 or -NO2; R3b is Y1; R3c is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), - S(O)2(ORx), -OC(Y1)Rx, -OC(Y1)ORx, -OC(Y1)(N(Rx)(Rx)), -SC(Y1)RX, - SC(Y!)ORx, -SC(Y1)(N(RX)(RX)), -N R^CQ^R*, -N(Rx)C(Y1)ORx, or - N(RX)C(Y1)(N(RX)(RX)) ; R3d is -C(Y1)RX, -C(Y*-)ORx or -C(YJ)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; W3 is W4 or W5; W4 is R5, -C(Y2)R5, -CC^^W5, -SOM2R5, or-SO^W5; W5 is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R2 groups; W6 is W3 independently substituted with 1, 2, or 3 A3 groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; and M12c is 0, 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11 or 12. In yet another embodiment the invention provides a kinase inhibiting conjugate that excludes such a compound. In another embodiment, the invention provides a compound ofthe formula: [DRUG]-(A°)nn or a pharmaceutically acceptable salt or solvate thereof wherein, DRUG is a compound of any one of formulae 500-511 (illustrated above); nn is 1, 2, or 3; A0 is A1, A2 or W3 with the proviso that the conjugate includes at least one A1; A1 is:
A2 is:
A3 is:
Y1 is independently O, S, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), or N(N(RX)( RX)); Y2 is independently a bond, O, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), N(N(RX)( Rx)), -S(O)M2-, or -S(O)M2-S(O)M2-; and when Y2 joins two phosphorous atoms Y2 can also be C(R2)(R2); Rx is independently H, R1, R2, W3, a protecting group, or the formula:
wherein: Ry is independently H, W3, R2 or a protecting group; R1 is independently H or alkyl of 1 to 18 carbon atoms; R2 is independently H, R1, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R3 groups or taken together at a carbon atom, two R2 groups form a ring of 3 to 8 carbons and the ring may be substituted with 0 to 3 R3 groups; R3 is R3a, R3b, R3c or R3d, provided that when R3 is bound to a heteroatom, then R3 is R3° or R3d; R3a is F, Cl, Br, I, -CN, N3 or -NO2; R3b is Y1; R30 is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), -
S(O)2(ORx), -OC(Y!)Rx, -OC(Y!)ORx, -OC(Y1)(N(Rx)(Rx)), -SC ^R", - SC ^OR*, -SC(Y1)(N(RX)(RX)), -N(RX)C(YJ)RX, -N(Rx)C(Y1)ORx, or - N(RX)C(Y1)(N(RX)(RX)) ; R3d is -C ^R", -C(Y1)ORx or -C(Y1)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; W3 is W4 or W5; W4 is R5, -C ^R5, -C(Y!)W5, -SO^R5, or-SO^W5; W5 is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R2 groups; W is W independently substituted with 1, 2, or 3 A groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; and M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12. In yet another embodiment the invention provides a kinase inhibiting conjugate that excludes such a compound. In another embodiment, the invention provides a compound of any one of formulae 1-36:
16 17
18 H
35 36
wherein: A0 is A1; A1 is:
A3 is:
Y1 is independently O, S, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), or N(N(RX)( RX)); Y2 is independently a bond, O, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), N(N(RX)( Rx)), -S(O)M2-, or -S(O)M2-S(O)M2-; and when Y2 joins two phosphorous atoms Y2 can also be C(R2)(R2); Rx is independently H, R2, W3, a protecting group, or the formula:
Ry is independently H, W3, R2 or a protecting group; R1 is independently H or alkyl of 1 to 18 carbon atoms; R2 is independently H, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R groups; RJ is R ,3a , r R,3b , R ,3icC or R ,3d , provided that when R' is bound to a heteroatom, then R3 is R3c or R3d; R3a is F, Cl, Br, I, -CN, N3 or -NO2; R3 is Y1; R3c is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), - S(O)2(ORx), -OC(Y-)Rx, -OC(Y1)ORx, -OC(Y1)(N(Rx)(Rx)), -SC(Y-)RX, SC(Y1)ORx, -SC(Y1)(N(RX)(RX)), -N(RX)C(Y1)RX, -N(Rx)C(Y1)ORx, or - N(RX)C(Y1)(N(RX)(RX)) ; R3d is -C(Y1)RX, -C(Y!)ORx or -C(YJ)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; R5a is independently alkylene of 1 to 18 carbon atoms, alkenylene of 2 to 18 carbon atoms, or alkynylene of 2-18 carbon atoms any one of which alkylene, alkenylene or alkynylene is substituted with 0-3 R3 groups; W3 is W4 or W5; W4 is R5, -CO W5, -SO2R5, or -SO2W5; W5 is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R2 groups; W6 is W3 independently substituted with 1, 2, or 3 A3 groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; X50 is H F, or Cl; and X51 is H or Cl. In yet another embodiment the invention provides a kinase inhibiting conjugate that excludes such a compound.
In another embodiment, the invention provides a compound of any one of formulae 500a-511a:
511a that is substituted with one or more groups A , wherein: A0 is A1, A2 or W3 with the proviso that the conjugate includes at least one A1; is:
A2 is:
A3 is: Y1 is independently O, S, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), or N(N(RX)( RX)); Y2 is independently a bond, O, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), N(N(RX)( Rx)), -S(O)M2-, or -S(O)M2-S(O)M2-; and when Y2 joins two phosphorous atoms Y2 can also be C(R2)(R2); Rx is independently H, R1, R2, W3, a protecting group, or the formula:
wherein: Ry is independently H, W3, R2 or a protecting group; R1 is independently H or alkyl of 1 to 18 carbon atoms; R2 is independently H, R1, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R groups or taken together at a carbon atom, two R groups form a ring of 3 to 8 carbons and the ring may be substituted with 0 to 3 R3 groups; R3 is R3a, R3 , R3c or R3d, provided that when R3 is bound to a heteroatom, then R3 is R3c or R3d; R3a is F, Cl, Br, I, -CN, N3 or -NO2; R^ is Y1; R3c is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), - S(O)2(ORx), -OC(Y1)Rx, -OC(Y1)ORx, -OC(Y1)(N(Rx)(Rx)), -SC(Y1)RX, - SCCY^OR3*, -SC(Y1)(N(RX)(RX)), -N(RX)C(Y1)RX, -N(Rx)C(Y1)ORx, or - N(RX)C(Y1)(N(RX)(RX)) ; R3d is -C(Y1)RX, -C(Y1)ORx or -C(Y1)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; W3 is W4 or W5; W4 is R5, -C ^R5, -C ^W5, -SO^R5, or-SO^W5; W5 is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R2 groups; W6 is W3 independently substituted with 1 , 2, or 3 A3 groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; and M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12. In yet another embodiment the invention provides a kinase inhibiting conjugate that excludes such a compound. In another embodiment, the invention provides a compound of any one of formulae la-36a:
21a 22a
H
29a 31a 32a
35a 36a
wherein: A0 is A1; A1 is:
A3 is:
Y1 is independently O, S, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), or N(N(RX)( Rx)); Y2 is independently a bond, O, N(RX), N(O)(Rx), N(ORx), N(O)(ORx), N(N(RX)( Rx)), -S(O)M2-, or -S(O)M2-S(O)M2-; and when Y2 joins two 9 9 9 phosphorous atoms Y can also be C(R )(R ); Rx is independently H, R2, W3, a protecting group, or the formula:
Ry is independently H, W3, R2 or a protecting group; R1 is independently H or alkyl of 1 to 18 carbon atoms; R2 is independently H, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R3 groups; R3 is R3a, R3b, R3c or R3d, provided that when R3 is bound to a heteroatom, then R3 is R3c or R3d; R3a is F, Cl, Br, I, -CN, N3 or -NO2; R3b is Y!; R3c is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), -
S(O)2(ORx), -OC(Y1)Rx, -OC(Y1)ORx, -OC(Y1)(N(Rx)(Rx)), -SC(Y1)RX, - SC(Y1)ORx, -SC(Y1)(N(RX)(RX)), -N(RX)C(Y1)RX, ^(R^C ^OR51, or - N(RX)C(Y1)(N(RX)(RX)) ; R3d is -C ^R", -C(YJ)ORx or -C(Y1)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; R5a is independently alkylene of 1 to 18 carbon atoms, alkenylene of 2 to 18 carbon atoms, or alkynylene of 2-18 carbon atoms any one of which alkylene, alkenylene or alkynylene is substituted with 0-3 R groups; W3 is W4 or W5; W4 is R5, -C ^R5, -C(Y1)W5, -SO2R5, or -SO2W5; W5 is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R groups; W6 is W3 independently substituted with 1 , 2, or 3 A3 groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; X50 is H F, or Cl; and X51 is H or Cl. In yet another embodiment the invention provides a kinase inhibiting conjugate that excludes such a compound.
Cellular Accumulation In one embodiment, the invention is provides compounds capable of accumulating in human PBMC (peripheral blood mononuclear cells). PBMC refer to blood cells having round lymphocytes and monocytes. Physiologically, PBMC are critical components ofthe mechanism against infection. PBMC may be isolated from heparinized whole blood of normal healthy donors or buffy coats, by standard density gradient centrifugation and harvested from the interface, washed (e.g. phosphate-buffered saline) and stored in freezing medium. PBMC may be cultured in multi-well plates. At various times of culture, supernatant may be either removed for assessment, or cells may be harvested and analyzed (Smith R. etal (2003) Blood 102(7):2532-2540). The compounds of this embodiment may further comprise a phosphonate or phosphonate prodrug. More typically, the phosphonate or phosphonate prodrug can have the structure A3 as described herein. Typically, compounds ofthe invention demonstrate improved intracellular half-life ofthe compounds or intracellular metabolites ofthe compounds in human PBMC when compared to analogs ofthe compounds not having the phosphonate or phosphonate prodrug. Typically, the half-life is improved by at least about 50%, more typically at least in the range 50-100%, still more typically at least about 100%, more typically yet greater than about 100%. In one embodiment ofthe invention the intracellular half-life of a metabolite ofthe compound in human PBMCs is improved when compared to an analog ofthe compound not having the phosphonate or phosphonate prodrug. In such embodiments, the metabolite may be generated intracellularly, e.g. generated within human PBMC. The metabolite may be a product ofthe cleavage of a phosphonate prodrug within human PBMCs. The phosphonate prodrug may be cleaved to form a metabolite having at least one negative charge at physiological pH. The phosphonate prodrug may be enzymatically cleaved within human PBMC to form a phosphonate having at least one active hydrogen atom of the form P-OH. Stereoisomers The compounds ofthe invention may have chiral centers, e.g. , chiral carbon or phosphorus atoms. The compounds ofthe invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers, and atropisomers. In addition, the compounds ofthe invention include enriched or resolved optical isomers at any or all asymmetric, chiral atoms. In other words, the chiral centers apparent from the depictions are provided as the chiral isomers or racemic mixtures. Both racemic and diastereomeric mixtures, as well as the individual optical isomers isolated or synthesized, substantially free of their enantiomeric or diastereomeric partners, are all within the scope ofthe invention. The racemic mixtures are separated into their individual, substantially optically pure isomers through well-known techniques such as, for example, the separation of diastereomeric salts formed with optically active adjuncts, e.g., acids or bases followed by conversion back to the optically active substances. In most instances, the desired optical isomer is synthesized by means of stereospecific reactions, beginning with the appropriate stereoisomer ofthe desired starting material. The compounds ofthe invention can also exist as tautomeric isomers in certain cases. All though only one delocalized resonance structure may be depicted, all such forms are contemplated within the scope ofthe invention. For example, ene-amine tautomers can exist for purine, pyrimidine, imidazole, guanidine, amidine, and tetrazole systems and all their possible tautomeric forms are within the scope ofthe invention. Salts and Hydrates The compositions of this invention optionally comprise salts ofthe compounds herein, especially pharmaceutically acceptable non-toxic salts containing, for example, Na+, Li+, K+> Ca+2 and Mg+2. Such salts may include those derived by combination of appropriate cations such as alkali and alkaline earth metal ions or ammonium and quaternary amino ions with an acid anion moiety, typically a carboxylic acid. Monovalent salts are preferred if a water soluble salt is desired. Metal salts typically are prepared by reacting the metal hydroxide with a compound of this invention. Examples of metal salts which are prepared in this way are salts containing Li+, Na+, and K+. A less soluble metal salt can be precipitated from the solution of a more soluble salt by addition ofthe suitable metal compound. In addition, salts may be formed from acid addition of certain organic and inorganic acids, e.g., HC1, HBr, H2SO45 H3PO4 or organic sulfonic acids, to basic centers, typically amines, or to acidic groups. Finally, it is to be understood that the compositions herein comprise compounds ofthe invention in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates. Also included within the scope of this invention are the salts ofthe parental compounds with one or more amino acids. Any ofthe amino acids described above are suitable, especially the naturally-occurring amino acids found as protein components, although the amino acid typically is one bearing a side chain with a basic or acidic group, e.g., Iysine, arginine or glutamic acid, or a neutral group such as glycine, serine, threonine, alanine, isoleucine, or leucine. Methods of Kinase Inhibition Another aspect ofthe invention relates to methods of inhibiting the activity of at least one kinase comprising the step of treating a sample suspected of containing a kinase with a composition ofthe invention. Compositions ofthe invention may act as kinase inhibitors, as intermediates for such inhibitors, or have other utilities as described herein. The inhibitors will bind to at least one kinase. Compositions binding the kinase may bind with varying degrees of reversibility. Those compounds binding substantially irreversibly are ideal candidates for use in this method ofthe invention. Once labeled, the substantially irreversibly binding compositions are useful as probes for the detection of a kinase. Accordingly, the invention relates to methods of detecting at least one kinase in a sample suspected of containing a kinase including the steps of: treating a sample suspected of containing kinase with a composition including a compound ofthe invention bound to a label; and observing the effect ofthe sample on the activity ofthe label. Suitable labels are well known in the diagnostics field and include stable free radicals, fluorophores, radioisotopes, enzymes, chemiluminescent groups and chromogens. The compounds herein are labeled in conventional fashion using functional groups such as hydroxyl or amino. Within the context ofthe invention, samples suspected of containing at least one kinase include natural or man-made materials such as living organisms; tissue or cell cultures; biological samples such as biological material samples (blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like. Typically the sample will be suspected of containing a kinase. Samples can be contained in any medium including water and organic solvent/water mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures. The treating step ofthe invention comprises adding the composition of the invention to the sample or it comprises adding a precursor ofthe composition to the sample. The addition step comprises any method of administration as described above. If desired, the activity of the kinase after application of the composition can be observed by any method including direct and indirect methods of detecting kinase activity. Quantitative, qualitative, and semiquantitative methods of determining kinase activity are all contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation ofthe physiological properties of a living organism are also applicable. Many organisms contain kinases. The compounds of this invention are useful in the treatment or prophylaxis of conditions associated with kinase activation in animals or in man. However, in screening compounds capable of inhibiting kinase it should be kept in mind that the results of enzyme assays may not correlate with cell culture assays. Thus, a cell based assay should be the primary screening tool. Screens for Kinase Inhibitors Compositions ofthe invention are screened for inhibitory activity against a kinase by any ofthe conventional techniques for evaluating enzyme activity. Within the context ofthe invention, typically compositions are first screened for inhibition of kinase in vitro and compositions showing inhibitory activity are then screened for activity in vivo. Compositions having in vitro Ki (inhibitory constants) of less then about 5 X 10"6 M, typically less than about 1 X 10~7 M and preferably less than about 5 X 10"8 M are preferred for in vivo use. Useful in vitro screens have been described, e.g., Bioorg. Med. Chem.
Pharmaceutical Formulations The compounds of this invention are formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986). Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH ofthe formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10. While it is possible for the active ingredients to be administered alone it may be preferable to present them as pharmaceutical formulations. The formulations, both for veterinary and for human use, ofthe invention comprise at least one active ingredient, as above defined, together with one or more acceptable carriers therefor and optionally other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients ofthe formulation and physiologically innocuous to the recipient thereof. The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any ofthe methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. Formulations ofthe present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount ofthe active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be administered as a bolus, electuary or paste. A tablet is made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture ofthe powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release ofthe active ingredient therefrom. For administration to the eye or other external tissues e.g. , mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1% and 20% in increments of 0.1% w/w such as 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w. When formulated in an ointment, the active ingredients may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase ofthe cream base may include, for example, at least 30% w/w of a polyhydric alcohol, t.e. an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3 -diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration ofthe active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulphoxide and related analogs. The oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase ofthe cream formulations. Emulgents and emulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate. The choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties. The cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils are used. Pharmaceutical formulations according to the present invention comprise one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed. Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil. Aqueous suspensions ofthe invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin. Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid. Dispersible powders and granules ofthe invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. The pharmaceutical compositions ofthe invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent. The pharmaceutical compositions ofthe invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% ofthe total compositions (weight: weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 μg ofthe active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur. Formulations suitable for administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w. Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier. Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate. Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions ofthe active ingredient. Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of conditions associated with kinase activity. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate. Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition ofthe sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets ofthe kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, ofthe active ingredient. It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents. The invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefor. Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally or by any other desired route. Compounds of the invention can also be formulated to provide controlled release ofthe active ingredient to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile ofthe active ingredient. Accordingly, the invention also provided compositions comprising one or more compounds ofthe invention formulated for sustained or controlled release. Effective dose of active ingredient depends at least on the nature ofthe condition being treated, toxicity, whether the compound is being used prophylactically (lower doses), the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day. Typically, from about 0.01 to about 10 mg/kg body weight per day. More typically, from about .01 to about 5 mg/kg body weight per day. More typically, from about .05 to about 0.5 mg/kg body weight per day. For example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from 1 mg to 1000 mg, preferably between 5 mg and 500 mg, and may take the form of single or multiple doses. Routes of Administration One or more compounds ofthe invention (herein referred to as the active ingredients) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient. An advantage ofthe compounds of this invention is that they are orally bioavailable and can be dosed orally. Combination Therapy Active ingredients ofthe invention are also used in combination with other active ingredients. Such combinations are selected based on the condition to be treated, cross-reactivities of ingredients and pharmaco-properties ofthe combination. It is also possible to combine any compound ofthe invention with one or more other active ingredients in a unitary dosage form for simultaneous or sequential administration to a patient. The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. The combination therapy may provide "synergy" and "synergistic effect", i.e. the effect achieved when the active ingredients used together is greater than the sum ofthe effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co- formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., in separate tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together. Metabolites ofthe Compounds ofthe Invention Also falling within the scope of this invention are the in vivo metabolic products ofthe compounds described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like ofthe administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabelled (e.g., C 4 or j 3) compound ofthe invention, administering it parenterally in a detectable dose (e.g. , greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g. , by MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing ofthe compounds ofthe invention even if they possess no kinase inhibitory activity of their own. Recipes and methods for detennining stability of compounds in surrogate gastrointestinal secretions are known. Compounds are defined herein as stable in the gastrointestinal tract where less than about 50 mole percent ofthe protected groups are deprotected in surrogate intestinal or gastric juice upon incubation for 1 hour at 37 °C. Simply because the compounds are stable to the gastrointestinal tract does not mean that they cannot be hydrolyzed in vivo. The phosphonate prodrugs ofthe invention typically will be stable in the digestive system but are substantially hydrolyzed to the parental drug in the digestive lumen, liver or other metabolic organ, or within cells in general. Exemplary Methods of Making the Compounds ofthe Invention. The invention also relates to methods of making the compositions ofthe invention. The compositions are prepared by any ofthe applicable techniques of organic synthesis. Many such techniques are well known in the art. However, many ofthe known techniques are elaborated in Compendium of Organic
Synthetic Methods (John Wiley & Sons, New York), Vol. 1, Ian T. Harrison and Shuyen Harrison, 1971; Vol. 2, Ian T. Harrison and Shuyen Harrison, 1974; Vol. 3, Louis S. Hegedus and Leroy Wade, 1977; Vol. 4, Leroy G. Wade, jr., 1980; Vol. 5, Leroy G. Wade, Jr., 1984; and Vol. 6, Michael B. Smith; as well as March, J., Advanced Organic Chemistrv. Third Edition. (John Wiley & Sons, New York, 1985), Comprehensive Organic Synthesis. Selectivity, Strategy & Efficiency in Modern Organic Chemistrv. In 9 Volumes, Barry M. Trost, Editor-in-Chief (Pergamon Press, New York, 1993 printing). A number of exemplary methods for the preparation ofthe compositions of the invention are provided below. These methods are intended to illustrate the nature of such preparations are not intended to limit the scope of applicable methods. Schemes and Examples General aspects of these exemplary methods are described below and in the Examples. Each ofthe products ofthe following processes is optionally separated, isolated, and/or purified prior to its use in subsequent processes. Generally, the reaction conditions such as temperature, reaction time, solvents, work-up procedures, and the like, will be those common in the art for the particular reaction to be performed. The cited reference material, together with material cited therein, contains detailed descriptions of such conditions. Typically the temperatures will be -100°C to 200°C, solvents will be aprotic or protic, and reaction times will be 10 seconds to 10 days. Work-up typically consists of quenching any unreacted reagents followed by partition between a water/organic layer system (extraction) and separating the layer containing the product. Oxidation and reduction reactions are typically carried out at temperatures near room temperature (about 20 °C), although for metal hydride reductions frequently the temperature is reduced to 0 °C to -100 °C, solvents are typically aprotic for reductions and may be either protic or aprotic for oxidations. Reaction times are adjusted to achieve desired conversions. Condensation reactions are typically carried out at temperatures near room temperature, although for non-equilibrating, kinetically controlled condensations reduced temperatures (0 °C to -100 °C) are also common. Solvents can be either protic (common in equilibrating reactions) or aprotic (common in kinetically controlled reactions). Standard synthetic techniques such as azeotropic removal of reaction by- products and use of anhydrous reaction conditions (e.g. , inert gas environments) are common in the art and will be applied when applicable. The terms "treated", "treating", "treatment", and the like, when used in connection with a chemical synthetic operation, mean contacting, mixing, reacting, allowing to react, bringing into contact, and other terms common in the art for indicating that one or more chemical entities is treated in such a manner as to convert it to one or more other chemical entities. This means that "treating compound one with compound two" is synonymous with "allowing compound one to react with compound two", "contacting compound one with compound two", "reacting compound one with compound two", and other expressions common in the art of organic synthesis for reasonably indicating that compound one was "treated", "reacted", "allowed to react", etc., with compound two. For example, treating indicates the reasonable and usual manner in which organic chemicals are allowed to react. Normal concentrations (0.01M to 10M, typically 0.1M to 1M), temperatures (-100 °C to 250 °C, typically -78 °C to 150 °C, more typically -78 °C to 100 °C, still more typically 0 °C to 100 °C), reaction vessels (typically glass, plastic, metal), solvents, pressures, atmospheres (typically air for oxygen and water insensitive reactions or nitrogen or argon for oxygen or water sensitive), etc., are intended unless otherwise indicated. The knowledge of similar reactions known in the art of organic synthesis are used in selecting the conditions and apparatus for "treating" in a given process. In particular, one of ordinary skill in the art of organic synthesis selects conditions and apparatus reasonably expected to successfully carry out the chemical reactions ofthe described processes based on the knowledge in the art. Modifications of each ofthe exemplary schemes and in the examples (hereafter "exemplary schemes") leads to various analogs ofthe specific exemplary materials produce. The above-cited citations describing suitable methods of organic synthesis are applicable to such modifications. In each ofthe exemplary schemes it may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium, and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography. Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like. Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like. Alternatively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like. Selection of appropriate methods of separation depends on the nature of the materials involved. For example, boiling point, and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply techniques most likely to achieve the desired separation. I l l A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution ofthe racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Stereochemistry of Carbon Compounds, (1962) by E. L. Eliel, McGraw Hill; Lochmuller, C. H., (1975) J. Chromatogr., 113:(3) 283-302). Racemic mixtures of chiral compounds ofthe invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods,
(2) formation of diastereomeric compounds with chiral derivatizing reagents, separation ofthe diastereomers, and conversion to the pure stereoisomers, and
(3) separation ofthe substantially pure or enriched stereoisomers directly under chiral conditions. Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, α-methyl-β-phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation ofthe optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation ofthe diastereomeric salts. Alternatively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. (1994) Stereochemistry of Organic Compounds. John Wiley & Sons, Inc., p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation ofthe diastereomers and hydrolysis to yield the free, enantiomerically enriched xanthene. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, α-methoxy-α- (trifluoromethyl)ρhenyl acetate (Jacob III. (1982) J. Org. Chem. 47:4165), ofthe racemic mixture, and analyzing the NMR spectrum for the presence ofthe two atropisomeric diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (Hoye, T., WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Chiral Liquid Chromatography (1989) W. J. Lough, Ed. Chapman and Hall, New York; Okamoto, (1990) J. ofChromatogr. 513:375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
Examples General Section A number of exemplary methods for the preparation of compounds ofthe invention are provided herein, for example, in the Examples hereinbelow. These methods are intended to illustrate the nature of such preparations are not intended to limit the scope of applicable methods. Certain compounds ofthe invention can be used as intermediates for the preparation of other compounds of the invention. For example, the interconversion of various phosphonate compounds ofthe invention is illustrated below. INTERCONVERSIONS OF THE PHOSPHONATES R-LINK-PfOVOR1 ?. R- LINK-P(O)(OR1 OH') AND R-UNK-P(O)(OH ?_. The following schemes 32-38 describe the preparation of phosphonate esters ofthe general structure R-link-P(O)(OR1)2, in which the groups R1 may be the same or different. The R1 groups attached to a phosphonate ester, or to precursors thereto, may be changed using established chemical transformations. The interconversion reactions of phosphonates are illustrated in Scheme S32. The group R in Scheme 32 represents the substructure, t.e. the drug "scaffold, to which the substituent link-P(O)(OR1)2 is attached, either in the compounds ofthe invention, or in precursors thereto. At the point in the synthetic route of conducting a phosphonate interconversion, certain functional groups in R may be protected. The methods employed for a given phosphonate transformation depend on the nature ofthe substituent R1, and ofthe substrate to which the phosphonate group is attached. The preparation and hydrolysis of phosphonate esters is described in Organic Phosphorus Compounds. G. M. Kosolapoff, L. Maeir, eds, Wiley, 1976, p. 9ff. In general, synthesis of phosphonate esters is achieved by coupling a nucleophile amine or alcohol with the corresponding activated phosphonate electrophilic precursor. For example, chlorophosphonate addition on to 5'- hydroxy of nucleoside is a well known method for preparation of nucleoside phosphate monoesters. The activated precursor can be prepared by several well known methods. Chlorophosphonates useful for synthesis ofthe prodrugs are prepared from the substituted- 1, 3 -propanediol (Wissner, et al, (1992) J. Med Chem. 35:1650). Chlorophosphonates are made by oxidation ofthe corresponding chlorophospholanes (Anderson, et al, (1984) J. Org. Chem. 49: 1304) which are obtained by reaction ofthe substituted diol with phosphorus trichloride. Alternatively, the chlorophosphonate agent is made by treating substituted-l,3-diols with phosphorusoxychloride (Patois, et al, (1990) J. Chem. Soc. Perkin Trans. 1, 1577). Chlorophosphonate species may also be generated in situ from corresponding cyclic phosphites (Silverburg, et al, (1996) Tetrahedron left., 31:111-11 A), which in turn can be either made from chlorophospholane or phosphoramidate intermediate. Phosphoroflouridate intermediate prepared either from pyrophosphate or phosphoric acid may also act as precursor in preparation of cyclic prodrugs (Watanabe et al, (1988) Tetrahedron lett., 29:5763-66). Phosphonate prodrugs ofthe present invention may also be prepared from the free acid by Mitsunobu reactions (Mitsunobu, (1981) Synthesis, 1; Campbell, (1992) J. Org. Chem. 57:6331), and other acid coupling reagents including, but not limited to, carbodiimides (Alexander, et al, (1994) Collect. Czech. Chem. Commun. 59:1853; Casara et al, (1992) Bioorg. Med. Chem. Lett. 2:145; Ohashi et al, (1988) Tetrahedron Lett, 29:1189), and ben-zotriazolyloxytris-(dimethylamino)phosphonium salts (Campagne et al (1993) Tetrahedron Lett. 34:6743). Aryl halides undergo Ni+2 catalyzed reaction with phosphite derivatives to give aryl phosphonate containing compounds (Balthazar, et al (1980) J. Org. Chem. 45:5425). Phosphonates may also be prepared from the chlorophosphonate in the presence of a palladium catalyst using aromatic triflates (Petrakis et al (1987) J. Am. Chem. Soc. 109:2831; Lu et al (1987) Synthesis 726). In another method, aryl phosphonate esters are prepared from aryl phosphates under anionic rearrangement conditions (Melvin (1981) Tetrahedron Lett 22:3375; Casteel et al (1991) Synthesis, 691). N-Alkoxy aryl salts with alkali met al derivatives of cyclic alkyl phosphonate provide general synthesis for heteroaryl-2-phosphonate linkers (Redmore (1970) J. Org. Chem. 35:4114). These above mentioned methods can also be extended to compounds where the W5 group is a heterocycle. Cyclic- 1, 3 -propanyl prodrugs of phosphonates are also synthesized from phosphonic diacids and substituted propane- 1 ,3 -diols using a coupling reagent such as 1 ,3 - dicyclohexylcarbodiimide (DCC) in presence of a base (e.g., pyridine). Other carbodiimide based coupling agents like 1,3-disopropylcarbodiimide or water soluble reagent, l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI) can also be utilized for the synthesis of cyclic phosphonate prodrugs. The conversion of a phosphonate diester S32.1 into the corresponding phosphonate monoester S32.2 (Scheme 32, Reaction 1) is accomplished by a number of methods. For example, the ester S32.1 in which R1 is an aralkyl group such as benzyl, is converted into the monoester compound S32.2 by reaction with a tertiary organic base such as diazabicyclooctane (DABCO) or quinuclidine, as described in J. Org*. Chem. (1995) 60:2946. The reaction is performed in an inert hydrocarbon solvent such as toluene or xylene, at about 110 °C. The conversion ofthe diester S32.1 in which R1 is an aryl group such as phenyl, or an alkenyl group such as allyl, into the monoester S32.2 is effected by treatment ofthe ester S32.1 with a base such as aqueous sodium hydroxide in acetonitrile or lithium hydroxide in aqueous tetrahydrofuran. Phosphonate diesters S32.1 in which one ofthe groups R1 is aralkyl, such as benzyl, and the other is alkyl, is converted into the monoesters S32.2 in which R1 is alkyl by hydrogenation, for example using a palladium on carbon catalyst. Phosphonate diesters in which both ofthe groups R1 are alkenyl, such as allyl, is converted into the monoester S32.2 in which R1 is alkenyl, by treatment with chlorotris(triphenylphosphine)rhodium (Wilkinson's catalyst) in aqueous ethanol at reflux, optionally in the presence of diazabicyclooctane, for example by using the procedure described in J. Org. Chem. (1973) 38:3224, for the cleavage of allyl carboxylates. The conversion of a phosphonate diester S32.1 or a phosphonate monoester S32.2 into the corresponding phosphonic acid S32.3 (Scheme 32, Reactions 2 and 3) can be effected by reaction ofthe diester or the monoester with trimethylsilyl bromide, as described in J. Chem. Soc, Chem. Comm., (1979) 739. The reaction is conducted in an inert solvent such as, for example, dichioromethane, optionally in the presence of a silylating agent such as bis(trimethylsilyl)trifluoroacetamide, at ambient temperature. A phosphonate monoester S32.2 in which R1 is aralkyl such as benzyl, is converted into the corresponding phosphonic acid S32.3 by hydrogenation over a palladium catalyst, or by treatment with hydrogen chloride in an ethereal solvent such as dioxane. A phosphonate monoester S32.2 in which R1 is alkenyl such as, for example, allyl, is converted into the phosphonic acid S32.3 by reaction with Wilkinson's catalyst in an aqueous organic solvent, for example in 15% aqueous acetonitrile, or in aqueous ethanol, for example using the procedure described in Helv. Chim. Acta. (1985) 68:618. Palladium catalyzed hydrogenolysis of phosphonate esters S32.1 in which R1 is benzyl is described in J. Org. Chem. (1959) 24:434. Platinum-catalyzed hydrogenolysis of phosphonate esters S32.1 in which R1 is phenyl is described in J. Am. Chem. Soc. (1956) 78:2336. The conversion of a phosphonate monoester S32.2 into a phosphonate diester S32.1 (Scheme 32, Reaction 4) in which the newly introduced R1 group is alkyl, aralkyl, haloalkyl such as chloroethyl, or aralkyl is effected by a number of reactions in which the substrate S32.2 is reacted with a hydroxy compound R^H, in the presence of a coupling agent. Typically, the second phosphonate ester group is different than the first introduced phosphonate ester group, i.e. R1 is followed by the introduction of R2 where each of R1 and R2 is alkyl, aralkyl, haloalkyl such as chloroethyl, or aralkyl (Scheme 32, Reaction 4a) whereby S32.2 is converted to S32.1a. Suitable coupling agents are those employed for the preparation of carboxylate esters, and include a carbodiimide such as dicyclohexylcarbodiimide, in which case the reaction is preferably conducted in a basic organic solvent such as pyridine, or (benzotriazol-1- yloxy)tripyrrolidinophosphonium hexafluorophosphate (PYBOP, Sigma), in wliich case the reaction is performed in a polar solvent such as dimethylformamide, in the presence of a tertiary organic base such as diisopropylethylamrne, or Aldrithiol-2 (Aldrich) in which case the reaction is conducted in a basic solvent such as pyridine, in the presence of a triaryl phosphine such as triphenylphosphine. Alternatively, the conversion ofthe phosphonate monoester S32.2 to the diester S32.1 is effected by the use ofthe Mitsunobu reaction, as described above (Scheme 7). The substrate is reacted with the hydroxy compound R*OH, in the presence of diethyl azodicarboxylate and a triarylphosphine such as triphenyl phosphine. Alternatively, the phosphonate monoester S32.2 is transformed into the phosphonate diester S32.1, in which the introduced R1 group is alkenyl or aralkyl, by reaction ofthe monoester with the halide R Br, in which R1 is as alkenyl or aralkyl. The alkylation reaction is conducted in a polar organic solvent such as dimethylformamide or acetonitrile, in the presence of a base such as cesium carbonate. Alternatively, the phosphonate monoester is transformed into the phosphonate diester in a two step procedure. In the first step, the phosphonate monoester S32.2 is transformed into the chloro analog RP(O)(OR1)Cl by reaction with thionyl chloride or oxalyl chloride and the like, as described in Organic Phosphorus Compounds, G. M. Kosolapoff, L. Maeir, eds, Wiley, 1976, p. 17, and the thus-obtained product RP(O)(OR1)Cl is then reacted with the hydroxy compound R^H, in the presence of a base such as triethylamine, to afford the phosphonate diester S32.1. A phosphonic acid R-link-P(O)(OH)2 is transformed into a phosphonate monoester RP(O)(OR1)(OH) (Scheme 32, Reaction 5) by means ofthe methods described above of for the preparation ofthe phosphonate diester R-link- P(O)(OR!)2 S32.1, except that only one molar proportion ofthe component R"OH or R*Br is employed. Dialkyl phosphonates may be prepared according to the methods of: Quast et al (1974) Synthesis 490; Stowell et al (1990) Tetrahedron Lett. 3261; US 5663159. A phosphonic acid R-link-P(O)(OH)2 S32.3 is transformed into a phosphonate diester R-link-P(O)(OR-) S32.1 (Scheme 32, Reaction 6) by a coupling reaction with the hydroxy compound R^H, in the presence of a coupling agent such as Aldrithiol-2 (Aldrich) and triphenylphosphine. The reaction is conducted in a basic solvent such as pyridine. Alternatively, phosphonic acids S32.3 are transformed into phosphonic esters S32.1 in which R1 is aryl, by means of a coupling reaction employing, for example, dicyclohexylcarbodiimide in pyridine at ca 70 "C. Alternatively, phosphonic acids S32.3 are transformed into phosphonic esters S32.1 in which R1 is alkenyl, by means of an alkylation reaction. The phosphonic acid is reacted with the alkenyl bromide R*Br in a polar organic solvent such as acetonitrile solution at reflux temperature, the presence of a base such as cesium carbonate, to afford the phosphonic ester S32.1. Scheme 32 O « R-link — Pχ-OR' R-link - -ii-OR1 OR1 , OH S32.2 S32.1 O R-link — R-OR1 R-link — P-OH OR1 OH S32.1 S32.3 O O I! R-link-P-OR1 **- R-link-P OH OH S32 2 OH S32.3 o o R-link— R-OR1 -* ^ R-link— PN-OR1 OH OR1 S32.1 S32.2
O O R-link— R-OR1 *- R-link— P OR1 OH OR2 S32.1a S32.2 O 5 O R-link — Pχ-OH ► R-llnk — Pχ-OR1 OH s32 3 OH S32.2
Preparation of phosphonate carbamates. Phosphonate esters may contain a carbamate linkage. The preparation of carbamates is described in Comprehensive Organic Functional Group Transformations, A. R. Katritzky, ed., Pergamon, 1995, Vol. 6, p. 416ff, and in Organic Functional Group Preparations, by S. R. Sandier and W. Karo, Academic Press, 1986, p. 260ff. The carbamoyl group may be formed by reaction of a hydroxy group according to the methods known in the art, including the teachings of Ellis, US 2002/0103378 Al and Hajima, US 6018049. Scheme 33 illustrates various methods by which the carbamate linkage is synthesized. As shown in Scheme 33, in the general reaction generating carbamates, an alcohol S33.1, is converted into the activated derivative S33.2 in which Lv is a leaving group such as halo, imidazolyl, benztriazolyl and the like, as described herein. The activated derivative S33.2 is then reacted with an amine S33.3, to afford the carbamate product S33.4. Examples 1 - 7 in Scheme 33 depict methods by which the general reaction is effected. Examples 8 - 10 illustrate alternative methods for the preparation of carbamates. Scheme 33, Example 1 illustrates the preparation of carbamates employing a chloroformyl derivative ofthe alcohol S33.5. In this procedure, the alcohol S33.5 is reacted with phosgene, in an inert solvent such as toluene, at about 0 °C, as described in Org. Svn. Coll. Vol. 3, 167, 1965, or with an equivalent reagent such as trichloromethoxy chloroformate, as described in Org. Svn. Coll. Vol. 6. 715, 1988, to afford the chloroformate S33.6. The latter compound is then reacted with the amine component S33.3, in the presence of an organic or inorganic base, to afford the carbamate S33.7. For example, the chloroformyl compound S33.6 is reacted with the amine S33.3 in a water- miscible solvent such as tetrahydrofuran, in the presence of aqueous sodium hydroxide, as described in Org. Syn. Coll. Vol. 3. 167, 1965, to yield the carbamate S33.7. Alternatively, the reaction is performed in dichioromethane in the presence of an organic base such as diisopropylethylamine or dimethylaminopyridine. Scheme 33, Example 2 depicts the reaction ofthe chloroformate compound S33.6 with imidazole to produce the imidazolide S33.8. The imidazolide product is then reacted with the amine S33.3 to yield the carbamate S33.7. The preparation ofthe imidazolide is performed in an aprotic solvent such as dichioromethane at 0°, and the preparation ofthe carbamate is conducted in a similar solvent at ambient temperature, optionally in the presence of a base such as dimethylaminopyridine, as described inJ. Med. Chem., 1989, 32, 357. Scheme 33 Example 3, depicts the reaction ofthe chloroformate S33.6 with an activated hydroxyl compound R"OH, to yield the mixed carbonate ester S33.10. The reaction is conducted in an inert organic solvent such as ether or dichioromethane, in the presence of a base such as dicyclohexylamine or triethylamine. The hydroxyl component R"OH is selected from the group of compounds S33.19 - S33.24 shown in Scheme 33, and similar compounds. For example, if the component R"OH is hydroxybenztriazole S33.19, N- hydroxysuccinimide S33.20, or pentachlorophenol, S33.21, the mixed carbonate S33.10 is obtained by the reaction ofthe chloroformate with the hydroxyl compound in an ethereal solvent in the presence of dicyclohexylamine, as described in Can. J. Chem., 1982, 60, 976. A similar reaction in which the component R"OH is pentafluorophenol S33.22 or 2-hydroxypyridine S33.23 is performed in an ethereal solvent in the presence of triethylamine, as described in Syn., 1986, 303, and Chem. Ber. 118, 468, 1985. Scheme 33 Example 4 illustrates the preparation of carbamates in which an alkyloxycarbonylimidazole S33.8 is employed. In this procedure, an alcohol S33.5 is reacted with an equimolar amount of carbonyl diimidazole S33.ll to prepare the intermediate S33.8. The reaction is conducted in an aprotic organic solvent such as dichioromethane or tetrahydrofuran. The acyloxyimidazole S33.8 is then reacted with an equimolar amount ofthe amine R'NH2 to afford the carbamate S33.7. The reaction is performed in an aprotic organic solvent such as dichioromethane, as described in Tet. Lett, 42, 2001, 5227, to afford the carbamate S33.7. Scheme 33, Example 5 illustrates the preparation of carbamates by means of an intermediate alkoxycarbonylbenztriazole S33.13. In this procedure, an alcohol ROH is reacted at ambient temperature with an equimolar amount of benztriazole carbonyl chloride S33.12, to afford the alkoxycarbonyl product S33.13. The reaction is performed in an organic solvent such as benzene or toluene, in the presence of a tertiary organic amine such as triethylamine, as described in Synthesis., 1977, 704. The product is then reacted with the amine R*NH2 to afford the carbamate S33.7. The reaction is conducted in toluene or ethanol, at from ambient temperature to about 80 °C as described in Synthesis., 1977, 704. Scheme 33, Example 6 illustrates the preparation of carbamates in which a carbonate (R"O)2CO, S33.14, is reacted with an alcohol S33.5 to afford the intermediate alkyloxycarbonyl intermediate S33.15. The latter reagent is then reacted with the amine R'NH2 to afford the carbamate S33.7. The procedure in which the reagent S33.15 is derived from hydroxybenztriazole S33.19 is described in Synthesis, 1993, 908; the procedure in which the reagent S33.15 is derived from N-hydroxysuccinimide S33.20 is described in Tet. Lett., 1992, 2781; the procedure in which the reagent S33.15 is derived from 2- hydroxypyridine S33.23 is described in Tet Lett, 1991, 4251; the procedure in which the reagent S33.15 is derived from 4-nitrophenol S33.24 is described in Synthesis. 1993, 103. The reaction between equimolar amounts ofthe alcohol ROH and the carbonate S33.14 is conducted in an inert organic solvent at ambient temperature. Scheme 33, Example 7 illustrates the preparation of carbamates from alkoxycarbonyl azides S33.16. In this procedure, an alkyl chloroformate S33.6 is reacted with an azide, for example sodium azide, to afford the alkoxycarbonyl azide S33.16. The latter compound is then reacted with an equimolar amount of the amine R'NH2 to afford the carbamate S33.7. The reaction is conducted at ambient temperature in a polar aprotic solvent such as dimethylsulfoxide, for example as described in Synthesis., 1982, 404. Scheme 33, Example 8 illustrates the preparation of carbamates by means ofthe reaction between an alcohol ROH and the chloroformyl derivative of an amine S33.17. In this procedure, which is described in Synthetic Organic Chemistrv. R. B. Wagner, H. D. Zook, Wiley, 1953, p. 647, the reactants are combined at ambient temperature in an aprotic solvent such as acetonitrile, in the presence of abase such as triethylamine, to afford the carbamate S33.7. Scheme 33, Example 9 illustrates the preparation of carbamates by means ofthe reaction between an alcohol ROH and an isocyanate S33.18. In this procedure, which is described in Synthetic Organic Chemistrv, R. B. Wagner, H. D. Zook, Wiley, 1953, p. 645, the reactants are combined at ambient temperature in an aprotic solvent such as ether or dichioromethane and the like, to afford the carbamate S33.7. Scheme 33, Example 10 illustrates the preparation of carbamates by means ofthe reaction between an alcohol ROH and an amine R'NH2. In this procedure, which is described in Chem. Lett. 1972, 373, the reactants are combined at ambient temperature in an aprotic organic solvent such as tefrahydrofuran, in the presence of a tertiary base such as triethylamine, and selenium. Carbon monoxide is passed through the solution and the reaction proceeds to afford the carbamate S33.7.
Scheme 33. Preparation of carbamates. General reaction R'NH2 ROH - ROCOLv ROCONHR S33.1 S33.2 S33.3 S33.4
Examples R'NH2 S33.3 (1) ROH- ROCOCI ROCONHR' S33.5 S33.6 S33.7
(2)
R'NH2 S33.3 ROCONHR' S33.7
R"OH R'NH2
(3) ROH *»- ROCOCI *- Kl JUUUK- ► KUL.UINI-1K S33.5 S33.6 S33.9 S33.10 S33.3 S33.7
R'
R" (R"02)C=0 R'NH, (6) ROH ROCOR" ROCONHR' S33.5 S33.14 S33.15 S33.3 S33.7
(7) ROH ROCOCI ROCON3 S33.5 S33.6 S33.16
R'NH233.3 ROCONHR' 33.7
(8) ROH R'NHCOCI ROCONHR' S33.5 S33.17 S33.7
R'NCO (9) ROH — — ► ROCONHR' S33.18 S33.5 S33.7
R'NH,
(10) ROH- ROCONHR' S33.5 S33.3 S33.7
PREPARATION OF CARBOALKOXY-SUBSTITUTED PHOSPHONATE BISAMIDATES. MONOAMIDATES. DIESTERS AND MONOESTERS. A number of methods are available for the conversion of phosphonic acids into amidates and esters. In one group of methods, the phosphonic acid is either converted into an isolated activated intermediate such as a phosphoryl chloride, or the phosphonic acid is activated in situ for reaction with an amine or a hydroxy compound. The conversion of phosphonic acids into phosphoryl chlorides is accomplished by reaction with thionyl chloride, for example as described in J. Gen. Chem. USSR, 1983, 53, 480, Zh. Obschei Khim., 1958, 28, 1063, or J. Org. Chem., 1994, 59, 6144, or by reaction with oxalyl chloride, as described in J. Am. Chem. Soc, 1994, 116, 3251, orJ. Org. Chem., 1994, 59, 6144, or by reaction with phosphorus pentachloride, as described inJ. Org. Chem., 2001, 66, 329, or inJ. Med. Chem., 1995, 38, 1372. The resultant phosphoryl chlorides are then reacted with amines or hydroxy compounds in the presence of a base to afford the amidate or ester products. Phosphonic acids are converted into activated imidazolyl derivatives by reaction with carbonyl diimidazole, as described in J. Chem. Soc, Chem. Comm. (1991) 312, or Nucleosides ά Nucleotides (2000) 19:1885. Activated sulfonyloxy derivatives are obtained by the reaction of phosphonic acids with trichloromethylsulfonyl chloride or with triisopropylbenzenesulfonyl chloride, as described in Tet. Lett. (1996) 7857, or Bioorg. Med. Chem. Lett. (1998) 8:663. The activated sulfonyloxy derivatives are then reacted with amines or hydroxy compounds to afford amidates or esters. Alternatively, the phosphonic acid and the amine or hydroxy reactant are combined in the presence of a diimide coupling agent. The preparation of phosphonic amidates and esters by means of coupling reactions in the presence of dicyclohexyl carbodiimide is described, for example, in J. Chem. Soc, Chem. Comm. (1991) 312 or Coll. Czech. Chem. Comm. (1987) 52:2792. The use of ethyl dimethylaminopropyl carbodiimide for activation and coupling of phosphonic acids is described in Tet Lett., (2001) 42:8841, ov Nucleosides & Nucleotides (2000) 19:1885. A number of additional coupling reagents have been described for the preparation of amidates and esters from phosphonic acids. The agents include Aldrithiol-2, and PYBOP and BOP, as described inJ. Org. Chem., 1995, 60, 5214, and J. Med. Chem. (1997) 40:3842, mesitylene-2-sulfonyl-3 -nitro- 1,2,4- triazole (MSNT), as described inJ. Med. Chem. (1996) 39:4958, diphenylphosphoryl azide, as described in J. Org. Chem. (1984) 49:1158, 1- (2,4,6-triisopropylbenzenesulfonyl-3-nitro-l,2,4-triazole (TPSNT) as described in Bioorg. Med. Chem. Lett. (1998) 8:1013, bromotris(dimethylamino)phosphonium hexafluorophosphate (BroP), as described in Tet. Lett, (1996) 37:3997, 2-chloro-5,5-dimethyl-2-oxo-l,3,2- dioxaphosphinane, as described in Nucleosides Nucleotides 1995, 14, 871, and diphenyl chlorophosphate, as described in J. Med. Chem., 1988, 31, 1305. Phosphonic acids are converted into amidates and esters by means ofthe Mitsunobu reaction, in which the phosphonic acid and the amine or hydroxy reactant are combined in the presence of a triaryl phosphine and a dialkyl azodicarboxylate. The procedure is described in Org. Lett, 2001, 3, 643, or J. Med. Chem., 1997, 40, 3842. Phosphonic esters are also obtained by the reaction between phosphonic acids and halo compounds, in the presence of a suitable base. The method is described, for example, in. Anal. Chem., 1987, 59, 1056, or J. Chem. Soc. Perkin Trans., I, 1993, 19, 2303, or J. Med. Chem., 1995, 38, 1372, or Tet Lett, 2002, 43, 1161. Schemes 34-37 illustrate the conversion of phosphonate esters and phosphonic acids into carboalkoxy-substituted phosphonbisamidates (Scheme 34), phosphonamidates (Scheme 35), phosphonate monoesters (Scheme 36) and phosphonate diesters, (Scheme 37). Scheme 38 illustrates synthesis of gem- dialkyl amino phosphonate reagents. Scheme 34 illustrates various methods for the conversion of phosphonate diesters S34.1 into phosphonbisamidates S34.5. The diester S34.1, prepared as described previously, is hydrolyzed, either to the monoester S34.2 or to the phosphonic acid S34.6. The methods employed for these transformations are described above. The monoester S34.2 is converted into the monoamidate S34.3 by reaction with an aminoester S34.9, in which the group R2 is H or alkyl; the group R is a divalent alkylene moiety such as, for example, CHCH3,
CHCH2CH3, CH(CH(CH3)2), CH(CH2Ph), and the like, or a side chain group present in natural or modified aminoacids; and the group R is C1-C12 alkyl, such as methyl, ethyl, propyl, isopropyl, or isobutyl; C6-C20 aryl, such as phenyl or substituted phenyl; or C6-C20 arylalkyl, such as benzyl or benzyhydryl. The reactants are combined in the presence of a coupling agent such as a carbodiimide, for example dicyclohexyl carbodiimide, as described in J. Am. Chem. Soc, (1957) 79:3575, optionally in the presence of an activating agent such as hydroxybenztriazole, to yield the amidate product S34.3. The amidate- forming reaction is also effected in the presence of coupling agents such as BOP, as described inJ. Org. Chem. (1995) 60:5214, Aldrithiol, PYBOP and similar coupling agents used for the preparation of amides and esters. Alternatively, the reactants S34.2 and S34.9 are transformed into the monoamidate S34.3 by means of a Mitsunobu reaction. The preparation of amidates by means ofthe Mitsunobu reaction is described inJ Med. Chem. (1995) 38:2742. Equimolar amounts of the reactants are combined in an inert solvent such as tetrahydrofuran in the presence of a triaryl phosphine and a dialkyl azodicarboxylate. The thus- obtained monoamidate ester S34.3 is then transformed into amidate phosphonic acid S34.4. The conditions used for the hydrolysis reaction depend on the nature ofthe R1 group, as described previously. The phosphonic acid amidate S34.4 is then reacted with an aminoester S34.9, as described above, to yield the bisamidate product S34.5, in which the amino substituents are the same or different. Alternatively, the phosphonic acid S34.6 may be treated with two different amino ester reagents simulataneously, i.e. S34.9 where R2, R or R5 are different. The resulting mixture of bisamidate products S34.5 may then be separable, e.g., by chromatography.
Scheme 34 O O O ii ^.,1 ιι . ■ R-link • ~PN-OR R-link - -OR1- R-link - -Pχ-OH 34.7 OR1 OH OH
R-link b)- C02R ,5b
S34.5
An example of this procedure is shown in Scheme 34, Example 1. In this procedure, a dibenzyl phosphonate S34.14 is reacted with diazabicyclooctane (DABCO) in toluene at reflux, as described in J. Org. Chem., 1995, 60, 2946, to afford the monobenzyl phosphonate S34.15. The product is then reacted with equimolar amounts of ethyl alaninate S34.16 and dicyclohexyl carbodiimide in pyridine, to yield the amidate product S34.17. The benzyl group is then removed, for example by hydrogenolysis over a palladium catalyst, to give the monoacid product S34.18 which may be unstable according to J. Med. Chem. (1997) 40(23):3842. This compound S34.18 is then reacted in a Mitsunobu reaction with ethyl leucinate S34.19, triphenyl phosphine and diethylazodicarboxylate, as described inJ Med. Chem., 1995, 38, 2742, to produce the bisamidate product S34.20. Using the above procedures, but employing in place of ethyl leucinate S34.19 or ethyl alaninate S34.16, different aminoesters S34.9, the corresponding products S34.5 are obtained. Alternatively, the phosphonic acid S34.6 is converted into the bisamidate S34.5 by use ofthe coupling reactions described above. The reaction is performed in one step, in which case the nitrogen-related substituents present in the product S34.5 are the same, or in two steps, in which case the nitrogen- related substituents can be different. An example ofthe method is shown in Scheme 34, Example 2. this procedure, a phosphonic acid S34.6 is reacted in pyridine solution with excess ethyl phenylalaninate S34.21 and dicyclohexylcarbodiimide, for example as described in J. Chem. Soc, Chem. Comm., 1991, 1063, to give the bisamidate product S34.22. Using the above procedures, but employing, in place of ethyl phenylalaninate, different aminoesters S34.9, the corresponding products S34.5 are obtained. As a further alternative, the phosphonic acid S34.6 is converted into the mono or bis-activated derivative S34.7, in which Lv is a leaving group such as chloro, imidazolyl, triisopropylbenzenesulfonyloxy etc. The conversion of phosphonic acids into chlorides S34.7 (Lv = Cl) is effected by reaction with thionyl chloride or oxalyl chloride and the like, as described in Organic Phosphorus Compounds, G. M. Kosolapoff, L. Maeir, eds, Wiley, 1976, p. 17. The conversion of phosphonic acids into monoimidazolides S34.7 (Lv = imidazolyl) is described inJ Med. Chem., 2002, 45, 1284 and inJ Chem. Soc. Chem. Comm., 1991, 312. Alternatively, the phosphonic acid is activated by reaction with triisopropylbenzenesulfonyl chloride, as described in Nucleosides and Nucleotides, 2000, 10, 1885. The activated product is then reacted with the aminoester S34.9, in the presence of a base, to give the bisamidate S34.5. The reaction is performed in one step, in which case the nitrogen substituents present in the product S34.5 are the same, or in two steps, via the intermediate S34.ll, in which case the nitrogen substituents can be different. Examples of these methods are shown in Scheme 34, Examples 3 and 5. In the procedure illustrated in Scheme 34, Example 3, a phosphonic acid S34.6 is reacted with ten molar equivalents of thionyl chloride, as described in Zh. Obschei Khim., 1958, 28, 1063, to give the dichloro compound S34.23. The product is then reacted at reflux temperature in a polar aprotic solvent such as acetonitrile, and in the presence of a base such as triethylamine, with butyl serinate S34.24 to afford the bisamidate product S34.25. Using the above procedures, but employing, in place of butyl serinate
S34.24, different aminoesters S34.9, the corresponding products S34.5 are obtained. In the procedure illustrated in Scheme 34, Example 5, the phosphonic acid S34.6 is reacted, as described in J Chem. Soc. Chem. Comm., 1991, 312, with carbonyl diimidazole to give the imidazolide S34.32. The product is then reacted in acetonitrile solution at ambient temperature, with one molar equivalent of ethyl alaninate S34.33 to yield the monodisplacement product S34.34. The latter compound is then reacted with carbonyl diimidazole to produce the activated intermediate S34.35, and the product is then reacted, under the same conditions, with ethyl N-methylalaninate S34.33a to give the bisamidate product S34.36. Using the above procedures, but employing, in place of ethyl alaninate S34.33 or ethyl N-methylalaninate S34.33a, different aminoesters S34.9, the corresponding products S34.5 are obtained. The intermediate monoamidate S34.3 is also prepared from the monoester S34.2 by first converting the monoester into the activated derivative S34.8 in which Lv is a leaving group such as halo, imidazolyl etc, using the procedures described above. The product S34.8 is then reacted with an aminoester S34.9 in the presence of a base such as pyridine, to give an intermediate monoamidate product S34.3. The latter compound is then converted, by removal ofthe R1 group and coupling ofthe product with the aminoester S34.9, as described above, into the bisamidate S34.5. An example of this procedure, in which the phosphonic acid is activated by conversion to the chloro derivative S34.26, is shown in Scheme 34, Example 4. In this procedure, the phosphonic monobenzyl ester S34.15 is reacted, in dichioromethane, with thionyl chloride, as described in Tet. Letters., 1994, 35, 4097, to afford the phosphoryl chloride S34.26. The product is then reacted in acetonitrile solution at ambient temperature with one molar equivalent of ethyl 3-amino-2-methylpropionate S34.27 to yield the monoamidate product S34.28. The latter compound is hydrogenated in ethylacetate over a 5% palladium on carbon catalyst to produce the monoacid product S34.29. The product is subjected to a Mitsunobu coupling procedure, with equimolar amounts of butyl alaninate S34.30, triphenyl phosphine, diethylazodicarboxylate and triethylamine in tetrahydrofuran, to give the bisamidate product S34.31. Using the above procedures, but employing, in place of ethyl 3-amino-2- methylpropionate S34.27 or butyl alaninate S34.30, different aminoesters S34.9, the corresponding products S34.5 are obtained. The activated phosphonic acid derivative S34.7 is also converted into the bisamidate S34.5 via the diamino compound S34.10. The conversion of activated phosphonic acid derivatives such as phosphoryl chlorides into the corresponding amino analogs S34.10, by reaction with ammonia, is described in Organic Phosphorus Compounds. G. M. Kosolapoff, L. Maeir, eds, Wiley, 1976. The bisamino compound S34.10 is then reacted at elevated temperature with a haloester S34.12 (Hal = halogen, i.e. F, Cl, Br, I), in a polar organic solvent such as dimethylformamide, in the presence of a base such as 4, 4- dimethylaminopyridine (DMAP) or potassium carbonate, to yield the bisamidate S34.5. Alternatively, S34.6 may be treated with two different amino ester reagents simulataneously, i.e. S34.12 where R4b or R5b are different. The resulting mixture of bisamidate products S34.5 may then be separable, e.g., by chromatography. An example of this procedure is shown in Scheme 34, Example 6. In this method, a dichlorophosphonate S34.23 is reacted with ammonia to afford the diamide S34.37. The reaction is performed in aqueous, aqueous alcoholic or alcoholic solution, at reflux temperature. The resulting diamino compound is then reacted with two molar equivalents of ethyl 2-bromo-3-mefhylbutyrate S34.38, in a polar organic solvent such as N-methylpyrrolidinone at ca. 150 °C, in the presence of a base such as potassium carbonate, and optionally in the presence of a catalytic amount of potassium iodide, to afford the bisamidate product S34.39. Using the above procedures, but employing, in place of ethyl 2-bromo-3- methylbutyrate S34.38, different haloesters S34.12 the corresponding products S34.5 are obtained. The procedures shown in Scheme 34 are also applicable to the preparation of bisamidates in which the aminoester moiety incorporates different functional groups. Scheme 34, Example 7 illustrates the preparation of bisamidates derived from tyrosine. In this procedure, the monoimidazolide S34.32 is reacted with propyl tyrosinate S34.40, as described in Example 5, to yield the monoamidate S34.41. The product is reacted with carbonyl diimidazole to give the imidazolide S34.42, and this material is reacted with a further molar equivalent of propyl tyrosinate to produce the bisamidate product S34.43. Using the above procedures, but employing, in place of propyl tyrosinate S34.40, different aminoesters S34.9, the corresponding products S34.5 are obtained. The aminoesters employed in the two stages ofthe above procedure can be the same or different, so that bisamidates with the same or different amino substituents are prepared. Scheme 35 illustrates methods for the preparation of phosphonate monoamidates. In one procedure, a phosphonate monoester S34.1 is converted, as described in Scheme 34, into the activated derivative S34.8. This compound is then reacted, as described above, with an aminoester S34.9, in the presence of a base, to afford the monoamidate product S35.1. The procedure is illustrated in Scheme 35, Example 1. In this method, a monophenyl phosphonate S35.7 is reacted with, for example, thionyl chloride, as described in J. Gen. Chem. USSR., 1983, 32, 367, to give the chloro product S35.8. The product is then reacted, as described in Scheme 34, with ethyl alaninate, to yield the amidate S35.10. Using the above procedures, but employing, in place of ethyl alaninate S35.9, different aminoesters S34.9, the corresponding products S35.1 are obtained. Alternatively, the phosphonate monoester S34.1 is coupled, as described in Scheme 34, with an aminoester S34.9 to produce the amidate S35.1. If necessary, the R1 substituent is then altered, by initial cleavage to afford the phosphonic acid S35.2. The procedures for this transformation depend on the nature ofthe R1 group, and are described above. The phosphonic acid is then transformed into the ester amidate product S35.3, by reaction with the hydroxy compound R3OH, in which the group R3 is aryl, heterocycle, alkyl, cycloalkyl, haloalkyl etc, using the same coupling procedures (carbodiimide, Aldrithiol-2, PYBOP, Mitsunobu reaction etc) described in Scheme 34 for the coupling of amines and phosphonic acids. Scheme 34 Example 1
R-link S34.14 S34.15 S34.1
Scheme 34 Example 2 Bn O H2NCH(Bn)CO2Et O V -COOEt
R-link - -i OH S34.21 R-link - -Pχ-NH OH NH Bn COOEt S34.6 S34.22 S34.25
Scheme 34 Example 4
S34.28
Scheme 34 Example 5
R-l
Scheme 34 Example 7
Examples of this method are shown in Scheme 35, Examples 1-3. In the sequence shown in Example 2, a monobenzyl phosphonate S35.ll is transformed by reaction with ethyl alaninate, using one ofthe methods described above, into the monoamidate S35.12. The benzyl group is then removed by catalytic hydrogenation in ethylacetate solution over a 5% palladium on carbon catalyst, to afford the phosphonic acid amidate S35.13. The product is then reacted in dichioromethane solution at ambient temperature with equimolar amounts of 1 -(dimethylaminopropyl)-3 -ethylcarbodiimide and trifluoroethanol S35.14, for example as described in Tet. Lett, 2001, 42, 8841, to yield the amidate ester S35.15. In the sequence shown in Scheme 35, Example 3, the monoamidate S35.13 is coupled, in tetrahydrofuran solution at ambient temperature, with equimolar amounts of dicyclohexyl carbodiimide and 4-hydroxy-N- methylpiperidine S35.16, to produce the amidate ester product S35.17. Using the above procedures, but employing, in place ofthe ethyl alaninate product S35.12 different monoacids S35.2, and in place of trifluoroethanol S35.14 or 4-hydroxy-N-methylρiperidine S35.16, different hydroxy compounds R3OH, the corresponding products S35.3 are obtained. Alternatively, the activated phosphonate ester S34.8 is reacted with ammonia to yield the amidate S35.4. The product is then reacted, as described in Scheme 34, with a haloester S35.5, in the presence of a base, to produce the amidate product S35.6. If appropriate, the nature ofthe R1 group is changed, using the procedures described above, to give the product S35.3. The method is illustrated in Scheme 35, Example 4. In this sequence, the monophenyl phosphoryl chloride S35.18 is reacted, as described in Scheme 34, with ammonia, to yield the amino product S35.19. This material is then reacted in N- methylpyrrolidinone solution at 170° with butyl 2-bromo-3-phenylproρionate S35.20 and potassium carbonate, to afford the amidate product S35.21. Using these procedures, but employing, in place of butyl 2-bromo-3- phenylpropionate S35.20, different haloesters S35.5, the corresponding products S35.6 are obtained. The monoamidate products S35.3 are also prepared from the doubly activated phosphonate derivatives S34.7. In this procedure, examples of which are described in Synlett, 1998, 1, 73, the intermediate S34.7 is reacted with a limited amount ofthe aminoester S34.9 to give the mono-displacement product S34.ll. The latter compound is then reacted with the hydroxy compound R OH in a polar organic solvent such as dimethylformamide, in the presence of a base such as diisopropylethylamine, to yield the monoamidate ester S35.3. The method is illustrated in Scheme 35, Example 5. In this method, the phosphoryl dichloride S35.22 is reacted in dichioromethane solution with one molar equivalent of ethyl N-methyl tyrosinate S35.23 and dimethylaminopyridine, to generate the monoamidate S35.24. The product is then reacted with phenol S35.25 in dimethylformamide containing potassium carbonate, to yield the ester amidate product S35.26. Using these procedures, but employing, in place of ethyl N-methyl tyrosinate S35.23 or phenol S35.25, the aminoesters S34.9 and/or the hydroxy compounds R OH, the corresponding products S35.3 are obtained. Scheme 35 S35.3
R-link S35.3
Scheme 35 Example 1
R-link
Scheme 35 Example 2
R-lin
O CF3CH2OH R-link — pχ-OCH2CF3 S35.14 NH Me— ( C02Et S35.15 Scheme 35 Example 3
S35.13 S35.17
Scheme 35 Example 4
O BrCH(Bn)C02Bu
R-link — R-OPh R-link - -PN-OPh R-link — -OPh Cl NH2 S35.20 NH Bn / S35.18 S35.19 C02Bu S35.21
Scheme 35 Example 5
R-lin
PhOH S35.25
Scheme 36 illustrates methods for the preparation of carboalkoxy- substituted phosphonate diesters in which one ofthe ester groups incorporates a carboalkoxy substituent. In one procedure, a phosphonate monoester S34.1, prepared as described above, is coupled, using one ofthe methods described above, with a hydroxyester S36.1, in which the groups R4b and R5b are as described in Scheme 34. For example, equimolar amounts ofthe reactants are coupled in the presence of a carbodiimide such as dicyclohexyl carbodiimide, as described in Aust J. Chem., 1963, 609, optionally in the presence of dimethylaminopyridine, as described in Tet, 1999, 55, 12997. The reaction is conducted in an inert solvent at ambient temperature. The procedure is illustrated in Scheme 36, Example 1. In this method, a monophenyl phosphonate S36.9 is coupled, in dichioromethane solution in the presence of dicyclohexyl carbodiimide, with ethyl 3-hydroxy-2- methylpropionate S36.10 to yield the phosphonate mixed diester S36.ll. Using this procedure, but employing, in place of ethyl 3-hydroxy-2- methylpropionate S36.10, different hydroxyesters S33.1, the corresponding products S33.2 are obtained. The conversion of a phosphonate monoester S34.1 into a mixed diester S36.2 is also accomplished by means of a Mitsunobu coupling reaction with the hydroxyester S36.1, as described in Org. Lett, 2001, 643. In this method, the reactants S34.1 and S36.1 are combined in a polar solvent such as tetrahydrofuran, in the presence of a triarylphosphine and a dialkyl azodicarboxylate, to give the mixed diester S36.2. The R1 substituent is varied by cleavage, using the methods described previously, to afford the monoacid product S36.3. The product is then coupled, for example using methods described above, with the hydroxy compound R OH, to give the diester product S36.4. The procedure is illustrated in Scheme 36, Example 2. In this method, a monoallyl phosphonate S36.12 is coupled in tetrahydrofuran solution, in the presence of triphenylphosphine and diethylazodicarboxylate, with ethyl lactate S36.13 to give the mixed diester S36.14. The product is reacted with tris(triphenylphosphine) rhodium chloride (Wilkinson catalyst) in acetonitrile, as described previously, to remove the allyl group and produce the monoacid product S36.15. The latter compound is then coupled, in pyridine solution at ambient temperature, in the presence of dicyclohexyl carbodiimide, with one molar equivalent of 3 -hydroxypyridine S36.16 to yield the mixed diester S36.17. Using the above procedures, but employing, in place ofthe ethyl lactate S36.13 or 3 -hydroxypyridine, a different hydroxyester S36.1 and/or a different hydroxy compound R3OH, the corresponding products S36.4 are obtained. The mixed diesters S36.2 are also obtained from the monoesters S34.1 via the intermediacy ofthe activated monoesters S36.5. In this procedure, the monoester S34.1 is converted into the activated compound S36.5 by reaction with, for example, phosphorus pentachloride, as described in J. Org: Chem., 2001 , 66, 329, or with thionyl chloride or oxalyl chloride (Lv *= Cl), or with triisopropylbenzenesulfonyl chloride in pyridine, as described in Nucleosides and Nucleotides, 2000, 19, 1885, or with carbonyl diimidazole, as described in J. Med. Chem., 2002, 45, 1284. The resultant activated monoester is then reacted with the hydroxyester S36.1, as described above, to yield the mixed diester S36.2. The procedure is illustrated in Scheme 36, Example 3. In this sequence, a monophenyl phosphonate S36.9 is reacted, in acetonitrile solution at 70 °C, with ten equivalents of thionyl chloride, so as to produce the phosphoryl chloride S36.19. The product is then reacted with ethyl 4-carbamoyl-2-hydroxybutyrate S36.20 in dichioromethane containing triethylamine, to give the mixed diester S36.21. Using the above procedures, but employing, in place of ethyl 4- carbamoyl-2-hydroxybutyrate S36.20, different hydroxyesters S36.1, the corresponding products S36.2 are obtained. The mixed phosphonate diesters are also obtained by an alternative route for incorporation ofthe R3O group into intermediates S36.3 in which the hydroxyester moiety is already incorporated. In this procedure, the monoacid intermediate S36.3 is converted into the activated derivative S36.6 in which Lv is a leaving group such as chloro, imidazole, and the like, as previously described. The activated intermediate is then reacted with the hydroxy compound R OH, in the presence of a base, to yield the mixed diester product S36.4. The method is illustrated in Scheme 36, Example 4. In this sequence, the phosphonate monoacid S36.22 is reacted with trichloromethanesulfonyl chloride in tetrahydrofuran containing collidine, as described inJ Med. Chem., 1995, 38, 4648, to produce the trichloromethanesulfonyloxy product S36.23. This compound is reacted with 3-(morpholinomethyl)phenol S36.24 in dichioromethane containing triethylamine, to yield the mixed diester product S36.25. Using the above procedures, but employing, in place of with 3- (morpholinomethyl)phenol S36.24, different alcohols R3OH, the corresponding products S36.4 are obtained. The phosphonate esters S36.4 are also obtained by means of alkylation reactions performed on the monoesters S34.1. The reaction between the monoacid S34.1 and the haloester S36.7 is performed in a polar solvent in the presence of a base such as diisopropylethylamine, as described in Anal. Chem., 1987, 59, 1056, or triethylamine, as described in J. Med. Chem., 1995, 38, 1372, or in a non-polar solvent such as benzene, in the presence of 18-crown-6, as described in Syn. Comm., 1995, 25, 3565. The method is illustrated in Scheme 36, Example 5. In this procedure, the monoacid S36.26 is reacted with ethyl 2-bromo-3 -phenylpropionate S36.27 and diisopropylethylamine in dimethylformamide at 80 °C to afford the mixed diester product S36.28. Using the above procedure, but employing, in place of ethyl 2-bromo-3- phenylpropionate S36.27, different haloesters S36.7, the corresponding products S36.4 are obtained.
Scheme 36
Hal-R4b-COOR5b S33.7
S36.6 S36.4
Scheme 36 Example 2
S36.14 S36.15
S36.16
Scheme 36 Example 3 O O F>-OPh SOCI
R-link - - R-link — p-OPh OH S36.18 Cl
S36.9 S36.19
Etθ2CCH
Scheme 36 Example 4
S36.22 S36.23
Scheme 36 Example 5
S36.26 S36.28
Scheme 37 illustrates methods for the preparation of phosphonate diesters in which both the ester substituents incoφorate carboalkoxy groups. The compounds are prepared directly or indirectly from the phosphonic acids S34.6. In one alternative, the phosphonic acid is coupled with the hydroxyester S37.2, using the conditions described previously in Schemes 34- 36, such as coupling reactions using dicyclohexyl carbodiimide or similar reagents, or under the conditions ofthe Mitsunobu reaction, to afford the diester product S37.3 in which the ester substituents are identical. This method is illustrated in Scheme 37, Example 1. In this procedure, the phosphonic acid S34.6 is reacted with three molar equivalents of butyl lactate S37.5 in the presence of Aldrithiol-2 and triphenyl phosphine in pyridine at ca. 70 °C, to afford the diester S37.6. Using the above procedure, but employing, in place of butyl lactate S37.5, different hydroxyesters S37.2, the corresponding products S37.3 are obtained. Alternatively, the diesters S37.3 are obtained by alkylation ofthe phosphonic acid S34.6 with a haloester S37.1. The alkylation reaction is performed as described in Scheme 36 for the preparation ofthe esters S36.4. This method is illustrated in Scheme 37, Example 2. In this procedure, the phosphonic acid S34.6 is reacted with excess ethyl 3-bromo-2- methylpropionate S37.7 and diisopropylethylamine in dimethylformamide at ca. 80 °C, as described in Anal Chem., 1987, 59, 1056, to produce the diester S37.8. Using the above procedure, but employing, in place of ethyl 3-bromo-2- methylpropionate S37.7, different haloesters S37.1, the corresponding products S37.3 are obtained. The diesters S37.3 are also obtained by displacement reactions of activated derivatives S34.7 ofthe phosphonic acid with the hydroxyesters S37.2. The displacement reaction is performed in a polar solvent in the presence of a suitable base, as described in Scheme 36. The displacement reaction is performed in the presence of an excess ofthe hydroxyester, to afford the diester product S37.3 in which the ester substituents are identical, or sequentially with limited amounts of different hydroxyesters, to prepare diesters S37.3 in which the ester substituents are different. The methods are illustrated in Scheme 37, Examples 3 and 4. As shown in Example 3, the phosphoryl dichloride S35.22 is reacted with three molar equivalents of ethyl 3-hydroxy-2-(hydroxymethyl)propionate S37.9 in tetrahydrofuran containing potassium carbonate, to obtain the diester product S37.10. Using the above procedure, but employing, in place of ethyl 3-hydroxy- 2-(hydroxymethyl)proρionate S37.9, different hydroxyesters S37.2, the corresponding products S37.3 are obtained. Scheme 37, Example 4 depicts the displacement reaction between equimolar amounts ofthe phosphoryl dichloride S35.22 and ethyl 2-methyl-3- hydroxypropionate S37.ll, to yield the monoester product S37.12. The reaction is conducted in acetonitrile at 70° in the presence of diisopropylethylamine. The product S37.12 is then reacted, under the same conditions, with one molar equivalent of ethyl lactate S37.13, to give the diester product S37.14. Using the above procedures, but employing, in place of ethyl 2-methyl-3- hydroxypropionate S37.ll and ethyl lactate S37.13, sequential reactions with different hydroxyesters S37.2, the corresponding products S37.3 are obtained.
Scheme 37
S34.7 S37.4 Scheme 37 Example 1 R-link
Scheme 37 Example 2
Scheme 37 Example 3
O (HOCH2)2CHCO2Et Q
R-hnk— P^-CI >- R |jnk — ^_0CH2CH(CH2OH)CO2Et S37.9 \ S35 22 OCH2CH(CH2OH)CO2Et S37.10 Scheme 37 Example 4 0 HOCH2CH(CH3)CO2Et O R-Hnk F»— Cl ***- R-link— P \-OCH2CH(CH3)CO2Et ci S37-11 Cl S37.12 S35.22
2,2-Dimethyl-2-aminoethylphosphonic acid intermediates can be prepared by the route in Scheme 38. Condensation of 2-methyl-2- propanesulfina ide with acetone give sulfinyl imine S38.ll (J. Org. Chem. 1999, 64, 12). Addition of dimethyl methylphosphonate lithium to S38.ll afford S38.12. Acidic methanolysis of S38.12 provide amine S38.13. Protection of amine with Cbz group and removal of methyl groups yield phosphonic acid S38.14, which can be converted to desired S38.15 (Scheme 38a) using methods reported earlier on. An alternative synthesis of compound S38.14 is also shown in Scheme 38b. Commercially available 2-amino-2-methyl-l-propanol is converted to aziridines S38.16 according to literature methods (J. Org. Chem. 1992, 57, 5813; Syn. Lett 1997, 8, 893). Aziridine opening with phosphite give S38.17 (Tetrahedron Lett 1980, 21, 1623). Reprotection of S38.17 affords S38.14. Scheme 38a
Scheme 38b
S38.14 The invention will now be illustrated by the following non-limiting
Examples. Example 1 Synthesis of Representative Compounds of Formulae 1-4
1.1 Representative compounds ofthe invention, e.g., as shown above, can be synthesized according to the following methods. CP-690,550 (3-{4-methyl-3-
[methyl-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-amino]-piperidin-l-yl}-3-oxo- propionitrile), can be prepared as described in WO 02/096,909 and WO
03/048,162. Enolate formation at the -cyanoamide position using over 2 equivalents of base followed by addition of diethyl phosphonomethyltriflate (prepared according to Tetrahedron Lett, 1986, 27, 1477) yields the desired compound 1.1 shown above. A solvent such as THF, DMF or other anhydrous solvents may be used for this reaction. In case the pyrrole nitrogen interferes with the desired alkylation, a protecting group such as BOC may be introduced before the alkylation reaction. Removal ofthe BOC group can be accomplished by exposure ofthe reaction product to TFA as described in Greene, T., Protective Groups In Organic Synthesis, Wiley-Interscience, 1999. Another specific compound ofthe invention can be synthesized as follows:
2A
(l-Benzyl-4-methyl-piρeridin-3-yl)-methyl-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- amine, compound 2.1 (prepared as described in WO 02/096,909) is first protected on the pyrrole nifrogen with a tosyl group. Subsequent formylation using the procedure reported by Sakamoto, T. et al, (Tetrahedron Lett. 1994, 35, 2919) provides compound 2.3. The primary alcohol is then treated in a solvent such as tetrahydrofriran or dimethylformamide with a base such as sodium hydride. When bubbling ceases, diethyl phosphonomethyltriflate (prepared according to Tetrahedron Lett, 1986, 27, 1477) is added, yielding the desired product 2.4. Debenzylation ofthe piperidine nitrogen following by coupling to cyano-acetic acid 2,5-dioxo-pyrrolidine-l-yl ester gives compound 2.5. Removal ofthe tosyl protecting group provides the desired compound 2A. Another specific compound ofthe invention can be synthesized as follows:
2-Amino-6-chloropurine is alkylated at the N-9 position by heating with 3-(t- butyldimethylsilyloxy)phenethyl iodide and sodium hydride in DMF, following a procedure similar to that described in US patent application 2002/0068721. The 2-amino group is converted to the iodo group by a conventional method such as that described in J. Med. Chem. 2003, 46, 5763. The resulting iodide is cross-coupled with cyclopentylzinc bromide in the presence of a palladium catalyst such as bis(triphenylphosphine)palladium(II) chloride (J. Org. Chem. 1991, 56, 1445). Transformation to the desired (4-{2-cycloρentyl-9-[2-(3- hydroxyphenyl)ethyl]-9H-purin-6-ylamino)phenoxymethyl)phosphonic acid diethyl ester is achieved by displacing the 6-chloro substituent with the corresponding phosphonate-containing aniline under reaction conditions such as those described in US patent application 2002/0068721, and then removing the t- butyldimethylsilyl protecting group by exposure to tetrabutylammonium fluoride. Another specific compound ofthe invention can be synthesized as follows:
A-420983 is demethylated by condensing with α-chloroethyl chloroformate in the presence of Hunig's base in a solvent such as chloroform, followed by brief heating in acidic methanol. The resulting free piperazine is alkylated with diethyl 2-bromoethylphosphonate in the presence of a base such as potassium carbonate, in a solvent such as dimethylformamide, to provide the desired product. All literature and patent citations herein are hereby expressly incorporated by reference at the locations of their citation. Specifically cited sections or pages ofthe above cited works are incorporated by reference with specificity. The invention has been described in detail sufficient to allow one of ordinary skill in the art to make and use the subject matter ofthe following embodiments. It is apparent that certain modifications ofthe methods and compositions ofthe following embodiments can be made within the scope and spirit ofthe invention. In the embodiments hereinbelow, the subscript and superscripts of a given variable are distinct. For example, Ri is distinct from R1.

Claims

Claims We claim: 1. A compound comprising one or more phosphonates and a substructure of formula I:
I wherein L1 and L2 are -N- or -CRa-; and Ra is hydrogen, alkyl, substituted alkyl, aryl or substituted aryl; or a pharmaceutically acceptable salt thereo.
2. The compound of claim 1 that comprises a substructure ofthe formula:
wherein: L1 and L2 are independently -N-, or -CRa-, provided that only one of L1 or L2 is a nitrogen atom; Ra is hydrogen, alkyl, aryl or substituted aryl; R20 is hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl aryl, cycloalkyl, substituted aryl, or -NRbR°; Rb and R° are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, or aralkyl; 91 R is hydrogen, alkyl, cycloalkyl, substituted cycloalkyl, substituted alkyl, aryl, substituted aryl, aralkyl, or substituted aralkyl; and R 1 2 and R ,23 are independently hydrogen, alkyl, substituted aryl, or aralkyl.
3. The compound of claim 1 that comprises a substructure of formula II:
π
4. The compound of claim 1 that comprises a substructure of formula Ilia, IVa or Va:
ffla IVa Va
5. The compound of claim 1 having formula 1, 2, 3, or 4:
wherein: A0 is A1; A1 is:
A3 is:
Y1 is independently O, S, N(RX), N(ORx), or N(N(RX)( Rx)); Y2 is independently a bond, O, N(RX), N(ORx), N(N(RX)( Rx)), or - S(O) 2-; and when Y2 joins two phosphorous atoms Y2 can also be C(R2)(R2); Rx is independently H, R2, W3, a protecting group, or the formula:
Ry is independently H, W3, R2 or a protecting group; R2 is independently H, R3 or R4 wherein each R4 is independently substituted with 0 to 3 R3 groups; R3 is R3a, R3b, R3c or R3d, provided that when R3 is bound to a heteroatom, then R3 is R3c or R3d: RR33aa iiss FF,, C Cl, Br, I, -CN, N3 or -NO2; R3b is Y1; R3° is -Rx, -N(RX)(RX), -SRX, -S(O)Rx, -S(O)2Rx, -S(O)(ORx), - S(O)2(ORx), -OC(Y1)Rx, -OC(Y1)ORx, -OC(Y1)(N(Rx)(Rx)), -SC(Y1)RX, - SC(Y1)ORx, -SC(Y1)(N(RX)(RX)), -N(RX)C(Y1)RX, -N(Rx)C(Y1)ORx, or - N(RX)C(Y!)(N(RX)(RX)) ; R3d is -CXY^R*, -C(Y1)ORx or -C(YJ)(N(RX)(RX)); R4 is an alkyl of 1 to 18 carbon atoms, alkenyl of 2 to 18 carbon atoms, or alkynyl of 2 to 18 carbon atoms; R5 is R4 wherein each R4 is substituted with 0 to 3 R3 groups; W3 is W4 or W5; W4 is R5, -CtY- R5, -C ^W5, -SO2R5, or -SO2W5; W is carbocycle or heterocycle wherein W5 is independently substituted with 0 to 3 R groups; W is W independently substituted with 1, 2, or 3 A groups; M2 is 0, 1 or 2; M12a is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; M12b is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; Mla, Mlc, and Mid are independently 0 or 1; M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; L1 and L2 are independently -N-, or -CRa-, provided that only one of L1 or L2 is a nitrogen atom; Ra is hydrogen, alkyl, aryl or substituted aryl; R20 is hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl aryl, cycloalkyl, substituted aryl, or -NRbR°; Rb and Rc are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, or aralkyl; 1 R is hydrogen, alkyl, cycloalkyl, substituted cycloalkyl, substituted alkyl, aryl, substituted aryl, aralkyl, or substituted aralkyl; and 99 R and R are independently hydrogen, alkyl, substituted aryl, or aralkyl.
6. The compound of claim 5 wherein A1 is ofthe formula:
7. The compound of claim 5 wherein A1 is ofthe formula:
The compound of claim 5 wherein A is ofthe formula
9. The compound of claim 5 wherein A1 is ofthe formula:
10. The compound of claim 5 wherein A1 is ofthe formula:
and W5a is a carbocycle or a heterocycle where W5a is independently substituted with 0 or 1 R2 groups.
11. The compound of claim 5 wherein M12a is 1.
12. The compound of claim 5 wherein A1 is ofthe formula:
13. The compound of claim 5 wherein A1 is ofthe formula:
14. The compound of claim 5 wherein A1 is ofthe formula:
W5a is a carbocycle independently substituted with 0 or 1 R2 groups;
15. The compound of claim 5 wherein A1 is ofthe formula:
Y2b is O orN(R2); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8.
16. The compound of claim 5 wherein A1 is ofthe formula:
1 R groups;
17. The compound of claim 5 wherein A1 is ofthe formula:
W5a is a carbocycle or heterocycle where W5a is independently substituted with 0 or 1 R2 groups.
18. The compound of claim 5 wherein A1 is of the formula:
Y2bisOorN(R2);and M12disl,2, 3,4, 5, 6, 7 or 8.
19. The compound of any one of claims 5-18 wherein A is of the formula:
20. The compound of any one of claims 5-18 wherein A is of the formula:
21. The compound of any one of claims 5-18 wherein A3 is of the formula: Y rlIadisOorS;and YaisO,N(Rx)orS.
22. The compound of any one of claims 5-18 wherein A 3 is of the formula:
andY r2bD :isOorN(Rx).
23. The compound of any one of claims 5-18 wherein A3 is of the formula:
R1 is independently H or alkyl of 1 to 18 carbon atoms; Y2bisOorN(Rx);and M12disl,2, 3,4, 5, 6, 7 or 8.
24. The compound of any one of claims 5-18 wherein A3 is of the formula: Y ^2D is O or N(Rx); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8.
25. The compound of claim 24 wherein Ml 2d is 1.
26. The compound of any one of claims 5-18 wherein A3 is of the formula:
27. The compound of any one of claims 5-18 wherein A is ofthe formula:
28. The compound of claim 27 wherein W is a carbocycle.
29. The compound of any one of claims 5-18 wherein A3 is of the formula:
30. The compound of claim 29 wherein W5 is phenyl.
31. The compound of claim 30 wherein M 12b is 1.
32. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
Yla is O or S; and Y r zaa is O, N(RX) or S.
33. The compound of any one of claims 5-18 wherein A is ofthe formula:
and Y r2ZbD is O or N(Rx).
34. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
R1 is independently H or alkyl of 1 to 18 carbon atoms; Y2b is O or N(Rx); and M12d is 1, 2, 3, 4, 5, 6, 7 or 8.
35. The compound of claim 34 wherein R1 is H.
36. The compound of claim 34 wherein M12d is 1.
37. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
wherein the phenyl carbocycle is substituted with 0, 1, 2, or 3 R2 groups.
38. The compound of any one of claims 5-18 wherein A3 is of the formula: wherein R1 is independently H or alkyl of 1 to 18 carbon atoms.
39. The compound of any one of claims 5-18 wherein A 3 is of the formula:
40. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
41. The compound of any one of claims 5-18 wherein A 3 is of the formula:
42. The compound of any one of claims 5-18 wherein A3 is of the formula:
Yla is O or S; and Y2aisO,N(R2)orS.
43. The compound of any one of claims 5-18 wherein A3 is of the formula:
Y rlιaa is O or S; Y2bisOorN(R2);and Y2c is O, N(Ry) or S.
44. The compound of any one of claims 5-18 wherein A3 is of the formula: R1 is independently H or alkyl of 1 to 18 carbon atoms; Y rlιaa is O or S; Y *isOorN(Ry);and M12d is 1,2, 3, 4, 5, 6, 7 or 8.
45. The compound of any one of claims 5-18 wherein A 3 is of the formula:
Y2bisOorN(R2);and M12d is 1,2, 3, 4, 5, 6, 7 or 8.
46. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
andY2bisOorN(R2).
47. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
48. The compound of any one of claims 5-18 wherein A is ofthe formula:
49. The compound of any one of claims 5-18 wherein A is of the formula:
Yla is O or S; and Y2a is O, N(R2) or S.
50. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
Yla is O or S; Y2bisOorN(R2);and Y2c is O, N(Ry) or S.
51. The compound of any one of claims 5-18 wherein A3 is of the formula:
R1 is independently H or alkyl of 1 to 18 carbon atoms; Yla is O or S; Y2bisOorN(R2); Y2disOorN(Ry);and M12disl,2,3,4, 5, 6, 7 or 8.
52. The compound of any one of claims 5-18 wherein A is of the formula:
Y2bisOorN(R2);and M12d is 1,2, 3, 4, 5, 6, 7 or 8.
53. The compound of any one of claims 5-18 wherein A3 is ofthe formula:
2b aαd Y D is O orN(Rz).
54. The compound of claim 5 wherein A0 is ofthe formula:
wherein each R is independently ( -C^alkyl.
55. The compound of claim 2 wherein: Ra is hydrogen, or substituted aryl; R20 is hydrogen, cycloalkyl, or -NRbRc; Rb is hydrogen, and Rc is substituted alkyl, or substituted aryl; 91 R is hydrogen, alkyl, substituted cycloalkyl, or substituted aralkyl; R22 is hydrogen, or alkyl; and R is hydrogen, substituted aryl, substituted cycloalkyl, or aralkyl.
56. The compound of any one of claims 1 -55 which inhibits a serine/threonine kinase, tyrosine kinase, Bcr-Abl kinase, cyclin-dependent kinase, Flt3 tyrosine kinase, MAP Erk kinase, JAK3 kinase, VEGF receptor kinase, PDGF receptor tyrosine kinase, protein kinase C, insulin receptor tyrosine kinase, and/or an EGF receptor tyrosine kinase.
57. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound as described in any one of claims 1-55.
58. A unit dosage form comprising a compound as described in any one of claims 1-55 and a pharmaceutically acceptable excipient.
59. A method for inhibiting a kinase in vitro or in vivo comprising contacting a sample in need of such treatment with a compound as described in any one of claims 1-55.
60. The method of claim 59 wherein the contacting is in vivo.
61. A method of inhibiting a kinase in an animal, comprising administering a compound as described in any one of claims 1-55 to the animal.
62. The method of claim 61 wherein the compound is formulated with a pharmaceutically acceptable carrier.
63. The method of claim 62 wherein the formulation further comprises a second active ingredient.
64. The method of any one of claims 59-63 wherein the kinase is a serine/threonine kinase, tyrosine kinase, Bcr-Abl kinase, cyclin-dependent kinase, Flt3 tyrosine kinase, MAP Erk kinase, JAK3 kinase, NEGF receptor kinase, PDGF receptor tyrosine kinase, protein kinase C, insulin receptor tyrosine kinase, and/or an EGF receptor tyrosine kinase.
65. A method of treating cancer in an animal in need of such treatment comprising administering an effective amount of a compound as described in any one of claims 1-55 to the animal.
66. A compound as described in any one of claims 1-55 for use in medical therapy.
67. The use of a compound as described in any one of claims 1 -55 to prepare a medicament for inhibiting a kinase in an animal.
68. The use of claim 67 wherein the kinase is a serine/threonine kinase, tyrosine kinase, Bcr-Abl kinase, cyclin-dependent kinase, Flt3 tyrosine kinase, MAP Erk kinase, JAK3 kinase, NEGF receptor kinase, PDGF receptor tyrosine kinase, protein kinase C, insulin receptor tyrosine kinase, and/or an EGF receptor tyrosine kinase.
69. The use of a compound as described in any one of claims 1 -55 to prepare a medicament for treating cancer in an animal.
70. A method for preparing a compound as described in the schemes and examples herein.
71. A method for preparing a pharmaceutical composition, comprising combining a pharmaceutically acceptable excipient and a compound as described in any one of claims 1-55.
EP04815625A 2003-12-22 2004-12-22 Kinase inhibitor phosphonate conjugates Withdrawn EP1715871A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US53193203P 2003-12-22 2003-12-22
US10/832,811 US7417055B2 (en) 2003-04-25 2004-04-26 Kinase inhibitory phosphonate analogs
PCT/US2004/013062 WO2004096234A2 (en) 2003-04-25 2004-04-26 Kinase inhibitor phosphonate conjugates
US62296204P 2004-10-26 2004-10-26
PCT/US2004/043595 WO2005063258A1 (en) 2003-12-22 2004-12-22 Kinase inhibitor phosphonate conjugates

Publications (1)

Publication Number Publication Date
EP1715871A1 true EP1715871A1 (en) 2006-11-02

Family

ID=34743875

Family Applications (1)

Application Number Title Priority Date Filing Date
EP04815625A Withdrawn EP1715871A1 (en) 2003-12-22 2004-12-22 Kinase inhibitor phosphonate conjugates

Country Status (6)

Country Link
EP (1) EP1715871A1 (en)
JP (1) JP2007529421A (en)
AU (1) AU2004308974A1 (en)
CA (1) CA2548951A1 (en)
MX (1) MXPA06007095A (en)
WO (1) WO2005063258A1 (en)

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG182849A1 (en) 2003-04-25 2012-08-30 Gilead Sciences Inc Antiviral phosphonate analogs
US7871991B2 (en) 2004-07-27 2011-01-18 Gilead Sciences, Inc. Phosphonate analogs of HIV inhibitor compounds
WO2006068760A2 (en) 2004-11-19 2006-06-29 The Regents Of The University Of California Anti-inflammatory pyrazolopyrimidines
US8546404B2 (en) 2005-12-13 2013-10-01 Merck Sharp & Dohme Compounds that are ERK inhibitors
KR20080103996A (en) 2006-02-16 2008-11-28 쉐링 코포레이션 Pyrrolidine derivatives as erk inhibitors
CA2647391C (en) 2006-04-04 2015-12-29 The Regents Of The University Of California Kinase antagonists
UA93548C2 (en) 2006-05-05 2011-02-25 Айерем Елелсі Compounds and compositions as hedgehog pathway modulators
GB2467670B (en) 2007-10-04 2012-08-01 Intellikine Inc Chemical entities and therapeutic uses thereof
EP3613743B1 (en) 2008-01-04 2022-03-16 Intellikine, LLC Processes for the preparation of 1h-pyrazolo[3,4-d]pyrimidin-4-amine derivatives
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
CA2714479A1 (en) 2008-02-21 2009-08-27 Schering Corporation Compounds that are erk inhibitors
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
WO2010006072A2 (en) 2008-07-08 2010-01-14 The Regents Of The University Of California Mtor modulators and uses thereof
CN102124009B (en) 2008-07-08 2014-07-23 因特利凯公司 Kinase inhibitors and methods of use
BRPI0915878A2 (en) 2008-07-08 2015-11-03 Gilead Sciences Inc salts or hydrates of HIV-inhibiting compounds, their use and pharmaceutical composition comprising them
SI2384326T1 (en) * 2008-08-20 2014-06-30 Zoetis Llc Pyrrološ2,3-dćpyrimidine compounds
EP2346508B1 (en) 2008-09-26 2016-08-24 Intellikine, LLC Heterocyclic kinase inhibitors
CA2740885C (en) 2008-10-16 2018-04-03 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
PE20100362A1 (en) 2008-10-30 2010-05-27 Irm Llc PURINE DERIVATIVES THAT EXPAND HEMATOPOYETIC STEM CELLS
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
CA2760791C (en) 2009-05-07 2017-06-20 Intellikine, Inc. Heterocyclic compounds and uses thereof
US7718662B1 (en) * 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
US8980899B2 (en) 2009-10-16 2015-03-17 The Regents Of The University Of California Methods of inhibiting Ire1
US8604032B2 (en) 2010-05-21 2013-12-10 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
WO2012064973A2 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
TWI674262B (en) 2011-01-10 2019-10-11 美商英菲尼提製藥股份有限公司 Processes for preparing isoquinolinones and solid forms of isoquinolinones
CA2828483A1 (en) 2011-02-23 2012-11-01 Intellikine, Llc Combination of kinase inhibitors and uses thereof
KR20140063605A (en) 2011-07-19 2014-05-27 인피니티 파마슈티칼스, 인코포레이티드 Heterocyclic compounds and uses thereof
AU2012284088B2 (en) 2011-07-19 2015-10-08 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
RU2014111823A (en) 2011-08-29 2015-10-10 Инфинити Фармасьютикалз, Инк. HETEROCYCLIC COMPOUNDS AND THEIR APPLICATIONS
MX370814B (en) 2011-09-02 2020-01-08 Univ California Substituted pyrazolo[3,4-d]pyrimidines and uses thereof.
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
CA2886240A1 (en) 2012-09-26 2014-04-03 The Regents Of The University Of California Modulation of ire1
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
HUE057734T2 (en) 2013-10-04 2022-06-28 Infinity Pharmaceuticals Inc Heterocyclic compounds and uses thereof
WO2015051241A1 (en) 2013-10-04 2015-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
CN106456628A (en) 2014-03-19 2017-02-22 无限药品股份有限公司 Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
WO2015160975A2 (en) 2014-04-16 2015-10-22 Infinity Pharmaceuticals, Inc. Combination therapies
WO2016054491A1 (en) 2014-10-03 2016-04-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
EP3350183A1 (en) 2015-09-14 2018-07-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinone derivatives, process of making, compositions comprising, and methods of using the same
WO2017161116A1 (en) 2016-03-17 2017-09-21 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as pi3k kinase inhibitors
WO2017214269A1 (en) 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
SG10201912456RA (en) 2016-06-24 2020-02-27 Infinity Pharmaceuticals Inc Combination therapies
EP3661937B1 (en) 2017-08-01 2021-07-28 Gilead Sciences, Inc. Crystalline forms of ethyl ((s)-((((2r,5r)-5-(6-amino-9h-purin-9-yl)-4-fluoro-2,5-dihydrofuran-2-yl)oxy)methyl)(phenoxy)phosphoryl)-l-alaninate (gs-9131) for treating viral infections

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5177064A (en) * 1990-07-13 1993-01-05 University Of Florida Targeted drug delivery via phosphonate derivatives
US5854227A (en) * 1994-03-04 1998-12-29 Hartmann; John F. Therapeutic derivatives of diphosphonates
US5874577A (en) * 1996-04-03 1999-02-23 Medichem Research, Inc. Method for the preparing 9-12-(Diethoxyphosphonomethoxy)ethyl!adenine and analogues thereof
CA2261619C (en) * 1996-07-26 2006-05-23 Gilead Sciences, Inc. Nucleotide analogs
JP2005523922A (en) * 2002-04-26 2005-08-11 ギリアード サイエンシーズ, インコーポレイテッド Non-nucleoside reverse transcriptase inhibitors
EP1620109A2 (en) * 2003-04-25 2006-02-01 Gilead Sciences, Inc. Kinase inhibitor phosphonate conjugates
WO2005002626A2 (en) * 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2005063258A1 *

Also Published As

Publication number Publication date
WO2005063258A1 (en) 2005-07-14
MXPA06007095A (en) 2006-09-04
CA2548951A1 (en) 2005-07-14
AU2004308974A1 (en) 2005-07-14
JP2007529421A (en) 2007-10-25

Similar Documents

Publication Publication Date Title
US7417055B2 (en) Kinase inhibitory phosphonate analogs
US9457035B2 (en) Antiviral compounds
US20070281907A1 (en) Kinase Inhibitor Phosphonate Conjugates
EP1715871A1 (en) Kinase inhibitor phosphonate conjugates
US20070072809A1 (en) Antiviral compounds
US20070027116A1 (en) Therapeutic phosphonate derivatives
US20070078081A1 (en) Antiviral compounds
US20080076740A1 (en) Antiviral compounds
WO2004096285A2 (en) Anti-infective phosphonate conjugates
US20090156558A1 (en) Phosphonate analogs of antimetabolites
WO2005012324A2 (en) Nucleobase phosphonate analogs for antiviral treatment
EP1809277A2 (en) Phosphonate derivatives of mycophenolic acid
US7427624B2 (en) Purine nucleoside phosphorylase inhibitory phosphonate compounds
US20050153990A1 (en) Phosphonate substituted kinase inhibitors
WO2006047507A2 (en) Phosphonate substituted kinase inhibitors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20060714

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1095767

Country of ref document: HK

17Q First examination report despatched

Effective date: 20070516

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090627

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1095767

Country of ref document: HK