WO2008057363A2 - Use of vitamin d derivatives to enhance delivery of therapeutics and oxygen to tumors - Google Patents

Use of vitamin d derivatives to enhance delivery of therapeutics and oxygen to tumors Download PDF

Info

Publication number
WO2008057363A2
WO2008057363A2 PCT/US2007/023030 US2007023030W WO2008057363A2 WO 2008057363 A2 WO2008057363 A2 WO 2008057363A2 US 2007023030 W US2007023030 W US 2007023030W WO 2008057363 A2 WO2008057363 A2 WO 2008057363A2
Authority
WO
WIPO (PCT)
Prior art keywords
vitamin
compound
mimic
animal
hydroxylase inhibitor
Prior art date
Application number
PCT/US2007/023030
Other languages
English (en)
French (fr)
Other versions
WO2008057363A3 (en
Inventor
John G. Curd
Original Assignee
Novacea, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novacea, Inc. filed Critical Novacea, Inc.
Publication of WO2008057363A2 publication Critical patent/WO2008057363A2/en
Publication of WO2008057363A3 publication Critical patent/WO2008057363A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/59Compounds containing 9, 10- seco- cyclopenta[a]hydrophenanthrene ring systems
    • A61K31/5929,10-Secoergostane derivatives, e.g. ergocalciferol, i.e. vitamin D2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention provides a method for preventing, treating or ameliorating hyperproliferative disorders and other conditions characterized by impaired performance status of an animal and/or inflammation by administering to the animal vitamin D compounds or a mimic thereof.
  • Vitamin D is a fat soluble vitamin which is essential as a positive regulator of calcium homeostasis. ⁇ See Harrison's Principles of Internal Medicine: Part Thirteen, "Disorders of Bone and Mineral Metabolism," Chapter 353, pp. 2214-2226, A.S. Fauci et al, (eds.), McGraw-Hill, New York (1998)).
  • the active form of vitamin D is l ⁇ ,25-dihydroxyvitamin D 3 , also known as calcitriol.
  • Specific nuclear receptors for active vitamin D compounds have been discovered in cells from diverse organs not involved in calcium homeostasis. Miller et al., Cancer Res. 52:515-520 (1992).
  • active vitamin D compounds have been implicated in osteogenesis, modulation of immune response, modulation of the process of insulin secretion by pancreatic B cells, muscle cell function, and the differentiation and growth of epidermal and hematopoietic tissues.
  • vitamin D compounds and analogues possess potent antileukemic activity by virtue of inducing the differentiation of malignant cells (specifically, leukemic cells) to non-malignant macrophages (monocytes) and are useful in the treatment of leukemia.
  • malignant cells specifically, leukemic cells
  • monocytes non-malignant macrophages
  • Vitamin D has been shown to inhibit the metastasis of lung cancer cells implanted in animals. Sato et al., Tohoku J. Exp. Med. ;j ⁇ 2:445-446 (1982).
  • Calcitriol was found to inhibit the growth of a lung cancer cell line containing high levels of vitamin D receptor but not lung cancer cell lines having insignificant levels of the receptor. Higashimoto et al, Anticancer Res. 76:2653-2660 (1996). Other reports suggesting important therapeutic uses of active vitamin D compounds are summarized in Rodriguez et al., U.S. Patent No. 6,034,074.
  • Active vitamin D compounds have also been administered in combination with other pharmaceutical agents, in particular cytotoxic agents for the treatment of hyperproliferative disease.
  • cytotoxic agents for the treatment of hyperproliferative disease.
  • pretreatment of hyperproliferative cells with active vitamin D compounds followed by treatment with cytotoxic agents enhances the efficacy of the cytotoxic agents (U.S. Patent Nos. 6,087,350; 6,559,139).
  • vitamin D compounds may result in substantial therapeutic benefits, the treatment of hyperproliferative diseases with such compounds is limited by the effects these compounds have on calcium metabolism.
  • active vitamin D compounds can induce markedly elevated and potentially dangerous blood calcium levels by virtue of their inherent calcemic activity. That is, the clinical use of calcitriol and other active vitamin D compounds as antiproliferative agents is severely limited by the risk of hypercalcemia.
  • the problem of systemic hypercalcemia can be overcome by "high dose pulse administration” (HDPA) of a sufficient dose of an vitamin D compound such that an antiproliferative effect is observed while avoiding the development of severe hypercalcemia.
  • HDPA high dose pulse administration
  • the active vitamin D compound may be administered no more than once every three days, for example, once a week at a dose of at least 0.12 ⁇ g/kg per day (8.4 ⁇ g in a 70 kg person).
  • compositions used in the HDPA regimen of 6,521,608 comprise 5-100 ⁇ g of active vitamin D compound and may be administered in the form for oral, intravenous, intramuscular, topical, transdermal, sublingual, intranasal, intratumoral or other preparations.
  • HDPA of calcitriol was shown to produce no dose-limiting toxicity and to produce mean peak calcitriol levels within the therapeutic range. Beer et al., Cancer 97:2431-39 (2001).
  • the invention provides a method for preventing, treating or ameliorating a hyperproliferative disorder or other condition in an animal characterized by diminished performance status, comprising administering to the animal a vitamin D compound or a mimic thereof, or a 24-hydroxylase inhibitor, preferably by high dose pulse administration.
  • the method comprises administering a 24-hydroxylase inhibitor in combination with a vitamin D compound or a mimic thereof.
  • the condition characterized by diminished performance status is a hyperproliferative disorder.
  • the condition is diabetes.
  • the condition is end stage kidney disease, e.g., a condition requiring dialysis.
  • the invention provides a method of treating a hyperproliferative disease or other condition in an animal comprising first determining the performance status of the animal and then administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24- hydroxylase inhibitor to the animal if the animal has been determined to exhibit diminished performance status.
  • the invention also provides a method of treating a hyperproliferative disease or other condition in an animal comprising first detecting a biomarker of inflammation in the animal and then administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to the animal if the animal has been determined to exhibit an elevated or diminished level of the biomarker.
  • the condition characterized by an elevated or diminished level of a biomarker of inflammation is a hyperproliferative disorder.
  • the condition is diabetes, hi yet another embodiment, the condition is end stage kidney disease, e.g., a condition requiring dialysis.
  • the invention also provides a method of improving delivery of a therapeutic agent to a tumor characterized by a reduced response to chemotherapy or radiation therapy, comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to the animal and then administering the therapeutic agent or radiation to the animal.
  • the invention also provides a method for improving the delivery of effector cells to a tumor in an animal, comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to said animal, thereby improving the anti-tumor immune response of the effector cells.
  • the invention also provides a method of improving delivery of a therapeutic agent to a tissue characterized by a reduced response to a therapeutic agent, comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to the animal and then administering the therapeutic agent to the animal.
  • the invention also provides a method of improving the perfusion of blood and oxygen to a tumor in an animal, comprising administering a vitamin D compound or mimic thereof to the animal, thereby improving the oxygenation of the tumor.
  • the administration leads to regulation of the tumor microenvironment, e.g., decreasing intratumoral pressure.
  • the administration promotes normalization of the tumor vasculature.
  • the increased oxygenation of the tumor due to the administration of a vitamin D compound or a mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor improves the response of the tumor to radiation therapy.
  • the invention also provides a method of improving the penetration and/or perfusion of a therapeutic agent into the tumor microenvironment relative to the tumor vasculature.
  • the treatment of an animal with a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor improves the limited penetration of a therapeutic agent into the tumor microenvironment to increase the therapeutic response in the animal.
  • the invention also provides a method of improving the perfusion of blood and oxygen to a tissue in an animal, comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to the animal, thereby improving the oxygenation of the tissue,
  • the administration leads to regulation of the tissue microenvironment, e.g., decreasing intratissue pressure.
  • the administration promotes normalization of the tissue vasculature.
  • the invention also provides a method of treating an animal with a tumor characterized by impaired perfusion and/or hypoxia, comprising determining whether the tumor exhibits impaired perfusion and/or hypoxia, administering to the animal a vitamin D compound or mimic thereof, a 24- hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor if the tumor has been determined to exhibit impaired perfusion and/or hypoxia, and administering to the tumor one or more radiotherapeutic agents or treatments.
  • the invention further provides a method of improving the response of a tumor to radiation therapy in an animal, comprising increasing oxygenation of the tumor by administering to the animal a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor.
  • the invention also provides a method of treating an animal with a condition characterized by impaired perfusion and/or hypoxia of a tissue, comprising determining whether the tissue exhibits impaired perfusion and/or hypoxia, administering to the animal a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor if the tissue has been determined to exhibit impaired perfusion and/or hypoxia, and administering to the tissue one or more therapeutic agents which are useful for treating said condition.
  • the invention also provides a method of treating an animal with a tumor characterized by impaired perfusion and/or hypoxia, comprising
  • the invention also provides a method of treating an animal with a condition characterized by impaired perfusion and/or hypoxia of a tissue, comprising
  • the invention also provides a method for palliative care of a terminal cancer patient, comprising administering to the patient a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor.
  • the vitamin D compound, or a mimic thereof is administered by high-dose pulse administration ("HDPA") so that high doses of the vitamin D compound or a mimic thereof, can be administered to an animal without inducing severe symptomatic hypercalcemia.
  • the vitamin D compound, or a mimic thereof is administered at a dose sufficient to obtain a peak plasma concentration of the vitamin D compound of at least 0.5 nM.
  • the vitamin D compound, or a mimic thereof is administered as a unit dosage form comprising about 10 ⁇ g to about 5 mg of calcitriol, about 50% MIGLYOL 812 and about 50% tocopherol PEG-1000 succinate (vitamin E TPGS).
  • the vitamin D compound, or a mimic thereof is administered as a unit dosage form comprising about 45 ⁇ g, about 90 ⁇ g, about 135 ⁇ g or about 180 ⁇ g.
  • the vitamin D compound or a mimic thereof may be administered orally, intravenously, parenterally, rectally, topically, nasally, sublingually, intramuscularly or transdermally.
  • Figure 1 Expected improvement of mitoxantrone penetration in mice pretreated with calcitriol compared to mitoxantrone penetration in control mice.
  • FIG. 1 The impact of an elevated baseline CRP on overall survival for men with metastatic androgen-independent prostate cancer (AIPC).
  • AIPC metastatic androgen-independent prostate cancer
  • the subgroup of patients with compromised baseline performance status demonstrated a diminished chemotherapeutic response to placebo and docetaxel as compared to those subjects with normal baseline performance status.
  • the diminished response to the docetaxel regimen in the subgroup with compromised baseline performance status is to be expected based on previous studies in AIPC and in other cancer tumor types with other chemotherapeutic regimens. See Jain, R.K., Science 307:58-62 (2005); Sessa, W.C., Cancer Cell (5/529-531 (2004).
  • the treatment of the subjects with compromised baseline performance status with calcitriol prior to the docetaxel chemotherapy regimen was associated with an increased response to docetaxel chemotherapy.
  • calcitriol administration enhanced the delivery and/or activity ⁇ i.e., increased the effectiveness) of docetaxel chemotherapy to the malignant tumors in patients with compromised baseline performance status.
  • One of the mechanisms that may explain these observations in part is the "normalization of tumor vasculature" by calcitriol via modulation of tumor vascular physiology. This is a potential mechanism of action for anti-VEGF treatment of tumors with chemotherapy.
  • Another putative mechanism is the suppression of inflammation within the tumor masses themselves leading to improved perfusion and delivery of the docetaxel chemotherapy that is administered intravenously.
  • calcitriol may decrease inflammation in the tumor which in turn decreases tumor interstitial fluid pressure and results in increased perfusion and/or distribution of chemotherapy agents into tumors, increased distribution of blood flow and oxygen to tumors, decreased vascular permeability and/or modulated tumor vessel architecture, thereby resulting in increased or more optimal chemotherapeutic delivery to the tumor microenvironment.
  • calcitriol and other vitamin D analogs and mimics, and agents that indirectly increases the level of vitamin D will be useful to enhance the delivery of chemotherapy and other agents to the tumors in patients with cancer who have an inadequate response to chemotherapy and other agents, thereby increasing the effectiveness of the chemotherapy.
  • calcitriol and other vitamin D analogs and mimics, and agents that indirectly increases the level of vitamin D will be useful to enhance the delivery of therapeutic agents to other tissues in patients having diminished performance status or in patients having a disease or disorder associated with inflammation, such as diabetes or kidney disease.
  • One embodiment of the present invention provides a method for preventing, treating or ameliorating hyperproliferative disorders and other conditions in animals characterized by diminished performance status, comprising administering to the animal a vitamin D compound or a mimic thereof, a 24-hydroxylase inhibitor, or a vitamin D compound or a mimic thereof in combination with a 24-hydroxylase inhibitor, hi another embodiment, the vitamin D compound or mimic thereof is administered by high dose pulse administration.
  • the performance status of the patient is determined using the ECOG performance status scale.
  • ECOG performance status refers to the ECOG scale and criteria used by doctors and researchers to assess how a disease affects the daily living abilities of the patient. ECOG performance status may be assessed according to Table 1. Table 1. ECOG Performance Status
  • animals are screened for ECOG performance status and those animals exhibiting an ECOG performance status of 1, 2, 3 or 4 are selected for further therapy with a vitamin D compound or mimic thereof.
  • the performance status of the patient is determined using the Karnofsky performance status scale.
  • Karnofsky performance status refers to the Karnofsky scale and criteria used by doctors and researchers to assess how a disease affects the daily living abilities of the patient.
  • Karnofsky performance status may be assessed according to Table 2.
  • animals are screened for Karnofsky performance status and those animals exhibiting a Karnofsky performance status of 80% or less are selected for further therapy with a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24- hydroxylase inhibitor.
  • the invention provides a method of treating a hyperproliferative disease or other condition in an animal by first determining the performance status of the animal and then administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to an animal that has been determined to exhibit diminished performance status.
  • hyperproliferative disease refers to any condition in which a localized population of proliferating cells in an animal is not governed by the usual limitations of normal growth, such as cancer.
  • hyperproliferative disorders include tumors, neoplasms, lymphomas and the like, as well as non-cancerous hyperproliferative conditions such as psoriasis.
  • a neoplasm is said to be benign if it does not undergo invasion or metastasis and malignant if it does either of these.
  • a "metastatic" cell means that the cell can invade and destroy neighboring body structures.
  • the invention provides a method of treating a hyperproliferative disease or other condition in an animal by first detecting a biomarker of inflammation in the animal and then administering a vitamin D compound or mimic thereof to an animal that has been determined to exhibit an elevated or diminished level of said biomarker.
  • the vitamin D compound or mimic thereof, 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor is administered if the level of said biomarker is elevated.
  • the method of treating a hyperproliferative disease or other condition comprises first detecting a biomarker of inflammation and detecting diminished performance status in an animal, and then administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to an animal that has been determined to exhibit an elevated or diminished level of the biomarker and a diminished performance status.
  • more than one biomarker of inflammation is detected, e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more biomarkers are detected.
  • a combination e.g., a predetermined set
  • biomarkers is detected.
  • the combination may be made up of biomarkers that are indicative of a particular type of inflammatory reaction or that are known to be predictive of certain outcomes of treatment.
  • the method of treatment may be used with animals having any disease or disorder that may be characterized by inflammation or thrombotic or other cardiovascular events.
  • the method of treatment is applied to animals with diabetes.
  • the method of treatment is applied to animals with end stage renal disease.
  • the method of treatment is applied to animals undergoing hemodialysis.
  • biomarker of inflammation refers to compounds, proteins, and other molecules that exhibit an increased or decreased level during an inflammatory response, such that detection of an increased or decreased level of the biomarker is indicative of the ongoing inflammatory response.
  • the elevated or diminished level of the biomarker may be detected in any bodily fluid (e.g., blood, serum, saliva, urine) or in tissue samples (e.g., biopsies).
  • biomarkers of inflammation include, without limitation, C-reactive protein (CRP), IL- l ⁇ , IL- l ⁇ , IL-4, IL-6, IL-8, IL-10, IL- 12, soluble IL-2 receptor, MCP-I, MMP-9, TNF ⁇ , soluble TNF receptor, fibrinogen, and IL-I receptor antagonist.
  • CRP C-reactive protein
  • IL- l ⁇ C-reactive protein
  • IL-4 IL-6
  • IL-8 IL-10
  • IL- 12 soluble IL-2 receptor
  • MCP-I MMP-9
  • TNF ⁇ soluble TNF receptor
  • the invention provides a method of improving delivery of a therapeutic agent to tumors characterized by a reduced response to chemotherapy or radiation therapy.
  • tumors may be characterized by abnormal vasculature which cause diminished perfusion and reduced delivery of the agent to the tumor.
  • a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor is administered to the animal and then the therapeutic agent or radiation is administered.
  • therapeutic agents include chemotherapeutic agents such as those described in more detail below, antibodies, protein and nucleic acid pharmaceuticals, and tumor targeted small molecular weight molecules.
  • the invention also provides a method of improving delivery of a therapeutic agent to a tissue characterized by a reduced response to a therapeutic agent.
  • tissue typically associated with the occurrence of a disease or disorder in an animal, may be characterized by inflammation or thrombotic or other cardiovascular events which cause diminished perfusion and reduced delivery of the agent to the tissue.
  • a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor is administered to the animal and then the therapeutic agent is administered.
  • reduced response to chemotherapy refers to a reduced response of a tumor to chemotherapeutic treatment as compared to the average response of the population at large.
  • reduced response to a therapeutic agent refers to a reduced response of a tissue to therapeutic treatment as compared to the average response of the population at large.
  • Chemotherapeutic agents useful in the invention include actinomycin
  • D irinotecan, vincristine, vinblastine, methotrexate, azathioprine, fluorouracil, doxorubicin, mitomycin, taxanes such as docetaxel, paclitaxel, and abraxane, cyclophosphamide, capecitabine, epirubicin, cisplatin, gemcitabine, mitoxantrone, leucovorin, vinorelbine, trastuzumab, etoposide, carboplatin, estramustine, prednisone, interferon alpha-2a, interleukin-2, bleomycin, ifosfamide, mesna, altretamine, topotecan, cytarabine, methylprednisolone, dexamethasone, daunorubicin, intrathecal methotrexate, mercaptopurine, thioguanine, fludarabine, gemtuzumab, i
  • the chemotherapeutic agent is a taxane, e.g., docetaxel or paclitaxel.
  • Therapeutic agents used for adjunctive or palliative treatment of cancer are also useful in the invention, including without limitation anti- emetics/anti-nausea agents (e.g., dolasetron, granisetron, ondansetron, tropisetron, palonosetron, domperidone, droperidol, haloperidol, chlorpromazine, promethazine, prochlorperazine, metoclopramide, alizapride, cyclizine, diphenhydramine, dimenhydrinate, meclizine, promethazine, hydroxyzine, cannabis, dronabinol, nabilone, sativex, midazolam, lorazepam, scopolamine) and mucositis prevention or treatment agents (e.g., allopurinol, benzydamine hydrochloride, cortic
  • Antibodies useful as therapeutic agents include, but are not limited to, monoclonal antibodies, synthetic antibodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv) (including bi-specific scFvs), single chain antibodies, Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), and epitope-binding fragments of any of the above.
  • antibodies include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds to an antigen.
  • the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGj, IgG 2 , IgG 3 , IgG 4 , IgA 1 and IgA 2 ) or subclass of immunoglobulin molecule.
  • the antibodies comprise four polypeptide chains-two light chains and two heavy chains, hi other embodiments, the antibodies of the invention comprise a V H chain and/or a V L chain. In yet another embodiments, the antibodies are epitope-binding fragments.
  • the antibodies may be from any animal origin including birds and mammals (e.g., human, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken).
  • the antibodies are human or humanized monoclonal antibodies.
  • "human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from mice or other animal that express antibodies from human genes.
  • the antibodies may be monospecific, bispecific, trispecific or of greater multispecificity.
  • Multispecific antibodies may immunospecifically bind to different epitopes of a polypeptide or may immunospecifically bind to both a polypeptide as well a heterologous epitope, such as a heterologous polypeptide or solid support material.
  • WO 93/17715, WO 92/08802, WO 91/00360, and WO 92/05793 Tutt, et ah, 1991, J. Immunol. 147:60-69; U.S. Pat. Nos. 4,474,893, 4,714,681, 4,925,648, 5,573,920, and 5,601,819; and Kostelny et al, 1992, J. Immunol. 148:1547- 1553.
  • the antibodies include derivatives of the antibodies known to those of skill in the art. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding an antibody to be used with the methods of the invention, including, for example, site- directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions.
  • the derivatives include less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original molecule.
  • the derivatives have conservative amino acid substitutions made at one or more predicted non-essential amino acid residues.
  • a "conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a side chain with a similar charge. Families of amino acid residues having side chains with similar charges have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed and the activity of the protein can be determined.
  • the antibodies include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, synthesis in the presence of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids.
  • one or more framework regions, preferably, all of the framework regions, of an antibody to be used in the methods of the invention or fragment thereof are human.
  • the fragment region of an antibody of the invention is humanized.
  • the antibody to be used with the methods of the invention is a synthetic antibody, a monoclonal antibody, an intrabody, a chimeric antibody, a human antibody, a humanized chimeric antibody, a humanized antibody, a glycosylated antibody, a multispecific antibody, a human antibody, a single-chain antibody, or a bispecific antibody.
  • the antibodies have half-lives in a mammal, preferably a human, of greater than 12 hours, greater than 1 day, greater than 3 days, greater than 6 days, greater than 10 days, greater than 15 days, greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months.
  • Antibodies or antigen-binding fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art.
  • antibodies or antigen-binding fragments thereof with increased in vivo half- lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fc domain and the FcRn receptor (see, e.g., PCT Publication No. WO 97/34631, U.S. patent application Ser. No. 10/020,354, entitled “Molecules with Extended Half-Lives, Compositions and Uses Thereof, filed Dec. 12, 2001, by Johnson et al., and U.S. patent application Ser. No. 11/263,230, filed Oct.
  • antibodies with increased in vivo half-lives can be generated by attaching to said antibodies or antibody fragments polymer molecules such as high molecular weight polyethyleneglycol (PEG).
  • PEG polymer molecules
  • PEG can be attached to said antibodies with or without a multifunctional linker either through site- specific conjugation of the PEG to the N- or C-terminus of said antibodies or via epsilon-amino groups present on lysine residues. Linear or branched polymer derivatization that results in minimal loss of biological activity will be used. The degree of conjugation will be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies.
  • Unreacted PEG can be separated from antibody-PEG conjugates by, e.g., size exclusion or ion-exchange chromatography.
  • PEG-derivatized antibodies or antigen-binding fragments thereof can be tested for binding activity to antigens as well as for in vivo efficacy using methods known to those skilled in the art, for example, by immunoassays described herein.
  • the antibodies can be single-chain antibodies.
  • the design and construction of a single-chain antibody is described in Marasco et al, 1993, Proc Natl Acad Sci 90:7889-7893, which is incorporated herein by reference in its entirety.
  • the antibodies bind to an intracellular epitope, i.e., are intrabodies.
  • An intrabody comprises at least a portion of an antibody that is capable of irnmunospecifically binding an antigen and preferably does not contain sequences coding for its secretion. Such antibodies will bind its antigen intracellularly.
  • the intrabody comprises a single- chain Fv ("sFv").
  • the intrabody does not encode an operable secretory sequence and thus remains within the cell (see generally Marasco, W A, 1998, “Intrabodies: Basic Research and Clinical Gene Therapy Applications” Springer: New York).
  • sFv are antibody fragments comprising the V H and V L domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains which enables the sFv to form the desired structure for antigen binding.
  • the antibodies may be conjugated or fused to one or more moieties, including but not limited to, peptides, polypeptides, proteins, fusion proteins, nucleic acid molecules, small molecules, mimetic agents, synthetic drugs, inorganic molecules, and organic molecules.
  • moieties including but not limited to, peptides, polypeptides, proteins, fusion proteins, nucleic acid molecules, small molecules, mimetic agents, synthetic drugs, inorganic molecules, and organic molecules.
  • the antibodies may also be recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to a heterologous protein or polypeptide (or fragment thereof, preferably to a polypeptide of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids) to generate fusion proteins.
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • antibodies may be used to target heterologous polypeptides to particular cell types, either in vitro or in vivo, by fusing or conjugating the antibodies to antibodies specific for particular cell surface receptors.
  • Antibodies fused or conjugated to heterologous polypeptides may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., International publication No. WO 93/21232; European Patent No. EP 439,095; Naramura et al, 1994, Immunol. Lett. 39:91-99; U.S. Pat. No. 5,474,981; Gillies et al, 1992, Proc. Natl. Acad. Sci. USA 89:1428-1432; and Fell et al, 1991, J. Immunol. 146:2446-2452, which are incorporated by reference in their entireties.
  • antibody fragments may be fused or conjugated to heterologous proteins, peptides or polypeptides.
  • the heterologous polypeptides may be fused or conjugated to a Fab fragment, Fd fragment, Fv fragment, F(ab) 2 fragment, a Fc domain, a VH domain, a VL domain, a VH CDR, a VL CDR, or fragment thereof.
  • Methods for fusing or conjugating polypeptides to antibody portions are well-known in the art. See, e.g., U.S. Pat. Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; European Patent Nos.
  • a fusion protein that comprises an Fc domain of an antibody or a fragment thereof may be used, wherein the Fc domain or the Fc domain fragment comprises at least one thioether cross-link.
  • a fusion protein can be any fusion protein comprising an Fc domain or an Fc domain fragment known in the art, such as human tumor necrosis factor receptor Fc fusion protein, as described in Moreland et al, 2000, New Eng. J. Med. 343:15869-93; or B7.1 Fc fusion protein, as described in Liu et al, 2005, Cancer Research l l(23):8492-8502, the contents of which are incorporated by reference in their entireties.
  • the Fc domain may further comprise one or more amino acid substitutions (Fc variants), hi some embodiments, Fc variants exhibit altered binding affinity for at least one or more Fc ligands ⁇ e.g., Fc ⁇ Rs, clq).
  • Fc variants and methods of making such are described for example, in U.S. Patent Publication Nos. 2006/0039904 and 2006/0040325, both published on Feb. 23, 2006, the contents of which are incorporated by reference in their entireties.
  • the fusion protein comprises a C H I, C H 2, C H 3 and/or C L domain of an antibody, wherein the C H I, C H 2, C H 3 or C L domain comprises at least one thioether cross-link.
  • DNA shuffling may be employed to alter the activities of antibodies or fragments thereof ⁇ e.g., antibodies or fragments thereof with higher affinities and lower dissociation rates). See, generally, U.S. Pat. Nos. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten et al, 1997, Curr. Opinion Biotechnol. 8:724-33; Harayama, 1998, Trends Biotechnol.
  • Antibodies or fragments thereof, or the encoded antibodies or fragments thereof may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination.
  • One or more portions of a polynucleotide encoding an antibody or antibody fragment may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules.
  • the antibodies can be conjugated to a diagnostic or detectable agent.
  • Such antibodies can be useful for monitoring or prognosing the development or progression of a disorder as part of a clinical testing procedure, such as determining the efficacy of a particular therapy.
  • Such diagnosis and detection can be accomplished by coupling the antibody to detectable substances including, but not limited to various enzymes, such as but not limited to horseradish peroxidase, alkaline phosphatase, beta- galactosidase, or acetylcholinesterase; prosthetic groups, such as but not limited to streptavidin/biotin and avidin/biotin; fluorescent materials, such as but not limited to, umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as but not limited to, luminol; bioluminescent materials, such as but not limited to, luciferase, luciferin, and aequorin; radioactive materials, such as but not limited to iodine ( 131 1, 125 I, 123 I, 1 21 I), carbon ( 14 C), sulfur ( 35 S
  • the present invention further encompasses antibodies that are conjugated to a therapeutic moiety.
  • An antibody or fragment thereof may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha- emitters.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Therapeutic moieties include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5- fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thiotepa chlorambucil, melphalan, carmustine (BCNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisdichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), Auristatin molecules (e.g
  • hormones e.g., glucocorticoids, progestins, androgens, and estrogens
  • DNA- repair enzyme inhibitors e.g., etoposide or topotecan
  • kinase inhibitors e.g., compound STl 571, imatinib mesylate (Kantaijian et al, Clin Cancer Res.
  • cytotoxic agents e.g., paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof) and those compounds disclosed in U.S. Pat. Nos.
  • antisense oligonucleotides e.g., those disclosed in the U.S. Pat. Nos. 6,277,832, 5,998,596, 5,885,834, 5,734,033, and 5,618,709
  • immunomodulators e.g., antibodies and cytokines
  • antibodies e.g., antibodies and cytokines
  • adenosine deaminase inhibitors e.g., Fludarabine phosphate and 2-Chlorodeoxyadenosine.
  • an antibody or fragment thereof may be conjugated to a therapeutic moiety or drug moiety that modifies a given biological response.
  • Therapeutic moieties or drug moieties are not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin; a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF ⁇ ., TNF ⁇ , AIM I (see, International publication No. WO 97/33899), AIM II (see, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, 1994, J.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue
  • a thrombotic agent or an anti-angiogenic agent e.g., angiostatin, endostatin or a component of the coagulation pathway (e.g., tissue factor); or, a biological response modifier such as, for example, a lymphokine (e.g., interleukin-1 ("IL-I”), interleukin-2 ("IL-2"), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), and granulocyte colony stimulating factor (“G-CSF”)), a growth factor (e.g., growth hormone (“GH”)), or a coagulation agent (e.g., calcium, vitamin K, tissue factors, such as but not limited to, Hageman factor (factor XII), high- molecular-weight kininogen (HMWK), prekallikrein (PK), coagulation proteins-factors
  • an antibody can be conjugated to therapeutic moieties such as a radioactive metal ion, such as alpha-emitters such as 213 Bi or macrocyclic chelators useful for conjugating radiometal ions, including but not limited to, 1 31 In, 131 LU, 131 Y, 131 Ho, 131 Sm, to polypeptides.
  • the macrocyclic chelator is 1, 4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraa- cetic acid (DOTA) which can be attached to the antibody via a linker molecule.
  • linker molecules are commonly known in the art and described in Denardo et al, 1998, Clin Cancer Res.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Pat. No. 4,676,980, which is incorporated herein by reference in its entirety.
  • the therapeutic moiety or drug conjugated to an antibody or fragment thereof should be chosen to achieve the desired prophylactic or therapeutic effect(s) for a particular disorder in a subject.
  • a clinician or other medical personnel should consider the following when deciding on which therapeutic moiety or drug to conjugate to an antibody or fragment thereof: the nature of the disease, the severity of the disease, and the condition of the subject.
  • the antibodies comprise at least one thioether cross-link, and wherein said antibodies specifically bind to one or more particular antigens.
  • the antibody of the present invention specifically binds to integrin ⁇ v ⁇ 3 .
  • the antibody comprises the amino acid sequence of the V H and V L chains of MEDI-522 (VitaxinTM). In other embodiments, the antibody comprises the amino acid sequence of the CDRs of the V H and V L chains of MEDI-522 (VitaxinTM).
  • the antibody of the present invention specifically binds to CD2.
  • the antibody comprises the amino acid sequence of the V H and V L chains of siplizumab.
  • the antibody comprises the amino acid sequence of the complementarity determining regions (CDRs) of V H and V L chains of siplizumab.
  • the antibody specifically binds to CDl 9. In some embodiments, the antibody comprises the amino acid sequence of the V H and V L chains of MT 103. In other embodiments, the antibody comprises the amino acid sequence of the CDRs of the V H and V L chains of MT 103. [0073] In certain embodiments, the antibody specifically binds to an Eph receptor. In certain embodiments, the antibody of the present invention specifically binds to EphA2. In some embodiments, the antibody comprises the amino acid sequence of the V H and V L chains of EA2 or EA5. In other embodiments, the antibody comprises the amino acid sequence of the CDRs of the V H and V L chains of EA2 or EA5. In certain embodiments, the antibody of the present invention specifically binds to EphA4. In some embodiments, the antibody of the present invention specifically binds to EphB4.
  • the antibody specifically binds to IL-9.
  • the antibody comprises the amino acid sequence of the V H and V L chains of MEDI-528.
  • the antibody comprises the amino acid sequence of the CDRs of the V H and V L chains of MEDI-528.
  • the antibody is RituxanTM (useful for treating non-Hodgkin's lymphoma), HerceptinTM (useful for treating metastatic breast cancer), CampathTM (useful for treating chronic lymphocytic leukemia), ErbituxTM (useful for treating various cancers), MDX-010 (useful for treating malignant melanoma, prostate cancer), MDX-214 (useful for treatment of cancer), AlloMuneTM (useful for treating non-Hodgkin's lymphoma, Hodgkin's disease), IMC-255 (antibody to epidermal growth factor), A7-neocarzinostatin (useful for the treatment of liver metastasis), 791T/36 (useful for the treatment of colorectal cancer), Fas/ APO-I (useful for treatment of malignant glioma cells), doxorubicin-CLNIgG (useful for the treatment of malignant glioma cells), siplizumab (useful
  • the antibody specifically binds to a protein involved in a disease, including, without limitation, EGFR, IGFR, HER2, VEGF, TNF, TGF, IL-4, IL-5, IL-8, IL-9, integrins, CD2, CD3, CD4, CDl Ia, CD14, CD18, CD19, CD20, CD22, CD23, CD25, CD33, CD40L, CD52, CD64, CD80, CD89, CD147, Eph receptor, CTLA4, GP Ilb/IIIa, C5, gpl20, VLA-4, ICAM-I, ICAM-3, E-selectin, Factor VII, HLA DR, TCR, CA 125, EpCAM, and IgE.
  • a protein involved in a disease including, without limitation, EGFR, IGFR, HER2, VEGF, TNF, TGF, IL-4, IL-5, IL-8, IL-9, integrins, CD2, CD3, CD4, CDl Ia, CD14
  • Therapeutic agents that may be used for the treatment of other conditions include, without limitation, analgesics, anti-inflammatory agents, anthelmintics, anti-arrhythmic agents, antibiotics, anticoagulants, antidepressants, antidiabetic agents, antiepileptics, antihistamines, antihypertensive agents, antimuscarinic agents, antimycobacterial agents, antineoplastic agents, immunosuppressants, antithyroid agents, antiviral agents, anxiolytic sedatives, astringents, beta-adrenoceptor blocking agents, cardiac inotropic agents, corticosteroids, cough suppressants, diuretics, dopaminergics, hemostatics, immunological agents, lipid regulating agents, muscle relaxants, parasympathomimetics, parathyroid calcitonin, prostaglandins, radiopharmaceuticals, sex hormones, anti-allergic agents, stimulants, anoretics, sympathomimetics, thyroid agents, vasodil
  • effector cells refers to immune system cells that exert the immune response in an individual.
  • effector cells include, without limitation, cells of monocyte/macrophage lineage, T cells (e.g., cytotoxic T cells), natural killer (NK) cells, peripheral blood mononuclear cells (PBMC), neutrophils, and mast cells.
  • T cells e.g., cytotoxic T cells
  • NK natural killer
  • PBMC peripheral blood mononuclear cells
  • neutrophils e.g., neutrophils, and mast cells.
  • the effector cells may be autologous or heterologous cells.
  • the effector cells may be modified to enhance their effectiveness, e.g., by increasing the expression of specific genes or exposing the cells to activators.
  • Isolating, modifying, and administering effector cells to animals may be carried out using techniques that are well known in the art.
  • the effector cells are autologous effector cells transformed with a gene coding for a cytokine, e.g., natural killer like T-lymphocytes transfected with a gene coding for interleukin-2 (IL-2). See Schmidt-Wolf, G.H. et al, Br. J. Cancer 57:1009-16 (1999).
  • the invention provides a method of treating cancer comprising administering a vitamin D compound or mimic thereof, a 24- hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to an animal in need thereof and then administering effector cells to the animal.
  • the invention provides a method of treating cancer comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to an animal in need thereof and then administering effector cells and a further therapeutic agent or treatment.
  • the invention provides a method of improving the perfusion of blood and oxygen to malignant tumors comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor, thereby improving the oxygenation of the tumors.
  • Oxygen is known to enhance the sensitivity malignant tumors to radiation treatment.
  • the invention also provides a method of treating an animal with malignant tumor(s) characterized by impaired perfusion and/or hypoxia, comprising determining whether the tumor exhibits impaired perfusion and/or hypoxia, administering to the animal a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor, and administering to the tumor(s) a radiotherapeutic agent or treatment, hi one embodiment, the administration of a vitamin D compound or a mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor regulates the tumor microenvironment, e.g., by decreasing intratumoral pressure.
  • Whether a tumor has hypoxic components and, thus, be amenable to treatment may be determined in a number of ways according to methods well known to those of ordinary skill in the art. For example one might insert an Eppendorff electrode into the tumor and compare the result obtained when the electrode is inserted into normal, perfused tissue. In another embodiment, the tumor may be imaged.
  • the invention also provides a method of improving the perfusion of blood and oxygen to a tissue in an animal, comprising administering a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor to the animal, thereby improving the oxygenation of the tissue.
  • tissue are typically associated with the occurrence of a disease or disorder in an animal.
  • the administration leads to regulation of the tissue microenvironment, e.g., decreasing intratissue pressure.
  • the administration promotes normalization of the tissue vasculature, hi a further embodiment, the increased oxygenation of the tumor due to the administration of a vitamin D compound or a mimic thereof, a 24- hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor improves the response of the tumor to radiation therapy.
  • the invention provides a method of improving the response of a tumor to radiation therapy in an animal, comprising increasing oxygenation of the tumor by administering to the animal a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor.
  • the invention also provides a method of improving the penetration and/or perfusion of a therapeutic agent into the tumor microenvironment relative to the tumor vasculature.
  • the treatment of a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24- hydroxylase inhibitor improves the limited penetration of a therapeutic agent into the tumor microenvironment to increase the therapeutic response in an animal.
  • the invention further provides that other agents may be coadministered which are known to "normalize” tumor or other tissue vasculature and enhance delivery of small organic molecules.
  • Such other agents include, without limitation, anti-VEGF and anti-VEGF receptor 2 antibodies.
  • the invention further provides that other agents may be coadministered that are known to decrease intratumoral or other intratissue pressure.
  • Such other agents include, without limitation, angiogenesis inhibitors such as anti-VEGF and anti-VEGF receptor 2 antibodies, anti-PDGF receptor antibodies, and conjugated TNP- 470.
  • coadministration refers to the simultaneous administration of two agents, either as a single composition or separate compositions.
  • the term also includes the administration of a first agent prior to or after the administration of a second agent, e.g., 1, 2, 3, 4, 5, 6, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks before or after the second agent.
  • the invention further provides that other agents may be coadministered which are known to decrease inflammation in tumors or other tissues.
  • Such other agents include corticosteroids (such as alclometasone, amcinonide, betamethasone, beclomethasone, budesonide, cortisone, clobetasol, clocortolone, desonide, dexamethasone, desoximetasone, diflorasone, flunisolide, fluticasone, fluocinonide, flurandrenolide, halcinonide, hydrocortisone, methylprednisolone, mometasone, prednicarbate, prednisone, prednisolone and triamcinolone), salicylates (such as aspirin, choline magnesium trisalicylate, methyl salicylate, salsalte and diflunisal), acetic acids (such as aspirin, choline magnesium trisalicylate,
  • the invention also provides a method of treating an animal with malignant tumor(s) characterized by impaired perfusion and/or hypoxia, comprising determining whether the tumor exhibits impaired perfusion and/or hypoxia, administering to the animal a vitamin D compound or mimic thereof and a compound known to decrease inflammation in tumors and administering to the tumor(s) one or more radiotherapeutic agents or treatments.
  • the invention also provides a method of treating an animal with a condition characterized by impaired perfusion and/or hypoxia of a tissue, comprising determining whether the tissue exhibits impaired perfusion and/or hypoxia, administering to the animal a vitamin D compound or mimic thereof, a 24-hydroxylase inhibitor, or the combination of a vitamin D compound or a mimic thereof and a 24-hydroxylase inhibitor and a compound known to decrease inflammation in tissues if the tissue has been determined to exhibit impaired perfusion and/or hypoxia, and administering to the tissue one or more therapeutic agents which are useful for treating said condition.
  • hypoxia refers to a deprivation of adequate oxygen supply.
  • Normoxia in human tissues other than the lung is about 6% (40 torr).
  • hypoxia is defined as a level of oxygen significantly less than 6%, e.g., 10, 20, 30, 40, 50, 60, 70, or 80% lower than 6%.
  • Hypoxia within a tumor or other tissue is defined as a level of oxygen within a portion or all of the tumor or other tissue that is at least 10% less than the level of oxygen in other tissues of the same individual, e.g., 20, 30, 40, 50, 60, 70, or 80% less.
  • the level of oxygen in a tissue may be determined by any method known in the art, such as oxygen electrodes (e.g., Eppendorf p ⁇ 2 Histograph), compounds that selectively bind to hypoxic regions (e.g., nitroimidazoles such as pimonidazole, and EF5), non-invasive techniques such as radiolabeled hypoxia markers that can be measured by positron emission tomography, single photon emission computed tomography, and magnetic resonance imaging, and techniques for measuring oxygen-dependent signals produced by the body's own chemistry, such as f-MRI BOLD (functional MRI blood oxygen level dependent), which detects magnetic properties of deoxyhemoglobin, recording variations in blood oxygen flow and tissue oxygen consumption, and NIRS (near-infrared spectroscopy), which measures the attenuation of light passing through tissue using an optical method to record oxygen-dependent signals from hemoglobin, deoxyhemoglobin, and cytochrome-c oxidase.
  • oxygen electrodes e.g., Epp
  • the term "impaired tissue perfusion and/or hypoxia induced by or associated with" one or more therapeutic agents refers to any impaired tissue perfusion and/or hypoxia that a patient develops during, or at the end of treatment with one or more therapeutic agents.
  • the term is intended to include any impaired tissue perfusion and/or hypoxia a patient suffers during or just after the end of the administration of one or more therapeutic agents (e.g., one or more chemotherapeutic agent, one or more radiotherapeutic agent, or combinations thereof) regardless of whether a direct or indirect causal link between the one or more therapeutic agents and the disorder can be demonstrated.
  • impaired tissue perfusion and/or hypoxia developed within about five weeks after the end of treatment with one or more therapeutic agents are included in "impaired tissue perfusion and/or hypoxia induced by or associated with” the one or more therapeutic agents.
  • impaired tissue perfusion and/or hypoxia that takes up to several months to develop after the end of treatment with the one or more therapeutic agents are included in "impaired tissue perfusion and/or hypoxia induced by or associated with” the one or more therapeutic agents.
  • the invention provides a method for palliative care of terminal cancer patients by administering to the patient a vitamin D compound or mimic thereof.
  • the term "palliative care,” as used herein, refers to the treatment of terminal patients to reduce the severity of their symptoms without curing the underlying disease, hi one embodiment, the method comprises administering a vitamin D compound or mimic thereof with one or more other agents that are known to be useful for palliative care, e.g., corticosteroids.
  • the vitamin D compound has a reduced hypercalcemic effect, allowing higher doses of the compound to be administered to an animal without inducing severe symptomatic hypercalcemia.
  • therapeutically effective amount refers to that amount of the therapeutic agent sufficient to achieve its intended result.
  • therapeutically effective amounts include those amounts which will improve the therapeutic response of another agent by at least 10%, preferably at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100%.
  • prevention refers to a decrease in the occurrence of pathological cells (e.g., hyperproliferative or neoplastic cells) in an animal.
  • the prevention may be complete, e.g., the total absence of pathological cells in a subject.
  • the prevention may also be partial, such that the occurrence of pathological cells in a subject is less than that which would have occurred without the present invention.
  • cancer is intended to refer to any known cancer, and may include, but is not limited to the following: leukemias such as acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblasts, promyelocytic, myelomonocytic, monocytic, and erythroleukemia leukemias, and myelodysplastic syndrome; chronic leukemias such as chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia; polycythemia vera; lymphomas such as Hodgkin's disease and non-Hodgkin's disease; multiple myelomas such as smoldering multiple myeloma, non-secretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma and extramedullary
  • Therapeutic agents useful as adjunctive therapy according to the invention include, but are not limited to, small molecules, synthetic drugs, peptides, polypeptides, proteins, nucleic acids (e.g., DNA and RNA polynucleotides including, but not limited to, antisense nucleotide sequences, triple helices, and nucleotide sequences encoding biologically active proteins, polypeptides, or peptides), antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules. Any agent which is known to be useful, or which has been used or is currently being used for the prevention, treatment, or amelioration of disorders can be used in combination with an vitamin D compound, or a mimic thereof, in accordance with the invention described herein.
  • nucleic acids e.g., DNA and RNA polynucleotides including, but not limited to, antisense nucleotide sequences, triple helices, and nucleotide sequences encoding biologically active proteins, polypeptides,
  • radiotherapeutic agent is intended to refer to any radiotherapeutic agent known to one of skill in the art to be effective to treat or ameliorate cancer, without limitation.
  • the radiotherapeutic agent can be an agent such as those administered in brachytherapy or radionuclide therapy.
  • Such methods can optionally further comprise the administration of one or more additional cancer therapies, such as, but not limited to, chemotherapies, surgery, and/or another radiotherapy.
  • a therapeutically effective dose of brachytherapy may be administered.
  • the brachytherapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation.
  • brachytherapy comprises insertion of radioactive sources into the body of a subject to be treated for cancer, preferably inside the tumor itself, such that the tumor is maximally exposed to the radioactive source, while preferably minimizing the exposure of healthy tissue.
  • the brachytherapy can be intracavitary brachytherapy.
  • the brachytherapy can be interstitial brachytherapy.
  • the brachytherapy can be administered at a high dose rate, a continuous low dose rate, or a pulsed dose rate.
  • a high dose rate brachytherapy regimen can be a dose of 60 Gy administered in ten fractions over six days
  • a continuous low dose rate brachytherapy regimen can be a total dose of about 65 Gy, administered continuously at about 40 to 50 cGy per hour.
  • Other examples of high, continuous low, and pulsed dose rate brachytherapy are well known in the art. See, e.g., Mazeron et al, Sem. Rad. One. 12:95-108 (2002).
  • Radioisotopes that can be administered in any of the above-described brachytherapies include, but are not limited to, phosphorus 32, cobalt 60, palladium 103, ruthenium 106, iodine 125, cesium 137, indium 192, xenon 133, radium 226, californium 252, or gold 198.
  • Other radioisotopes may be selected for administration in brachytherapy according to the desirable physical properties of such a radioisotope.
  • Radioisotope's suitability for use in brachytherapy including, but not limited to, the radioisotope's half-life, the degree to which emitted radiation penetrates surrounding tissue, the energy of emitted radiation, the ease or difficulty of adequately shielding the radioisotope, the availability of the radioisotope, and the ease or difficulty of altering the shape of the radioisotope prior to administration.
  • the vitamin D compound, or a mimic thereof may be administered in combination with a treatment comprising a therapeutically effective dose of a radionuclide.
  • the radionuclide therapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation, hi general, radionuclide therapy comprises systemic administration of a radioisotope that preferentially accumulates in or binds to the surface of cancerous cells.
  • the preferential accumulation of the radionuclide can be mediated by a number of mechanisms, including, but not limited to, incorporation of the radionuclide into rapidly proliferating cells, specific accumulation of the radionuclide by the cancerous tissue without special targeting (e.g., iodine 131 accumulation in thyroid cancer), or conjugation of the radionuclide to a biomolecule specific for a neoplasm.
  • mechanisms including, but not limited to, incorporation of the radionuclide into rapidly proliferating cells, specific accumulation of the radionuclide by the cancerous tissue without special targeting (e.g., iodine 131 accumulation in thyroid cancer), or conjugation of the radionuclide to a biomolecule specific for a neoplasm.
  • a specific biomolecule for use in targeting a particular neoplasm for radionuclide therapy based upon the cell-surface molecules present on that neoplasm.
  • hepatomas may be specifically targeted by an antibody specific for ferritin, which is frequently over-expressed in such tumors.
  • antibody-targeted radioisotopes for the treatment of cancer include ZEVALIN (ibritumomab tiuxetan) and BEXXAR (tositumomab), both of which comprise an antibody specific for the B cell antigen CD20 and are used for the treatment of non-Hodgkin lymphoma.
  • radiotherapeutic treatment is intended to refer to any radiotherapeutic treatment known to one of skill in the art to be effective to treat or ameliorate cancer, without limitation.
  • the radiotherapeutic treatment can be external-beam radiation therapy, thermotherapy, radiosurgery, charged-particle radiotherapy, neutron radiotherapy, or photodynamic therapy.
  • Such methods can optionally further comprise the administration of one or more additional cancer therapies, such as, but not limited to, chemotherapies, surgery, and/or another radiotherapy.
  • the present invention provides a method comprising the administration of a vitamin D compound, or a mimic thereof, in combination with a treatment comprising a therapeutically effective dose of external-beam radiation therapy.
  • the external-beam radiation therapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation.
  • external-beam radiation therapy comprises irradiating a defined volume within a subject with a high energy beam, thereby causing cell death within that volume.
  • the irradiated volume preferably contains the entire cancer to be treated, and preferably contains as little healthy tissue as possible.
  • the external-beam radiation therapy can be three-dimensional conformal radiotherapy.
  • the external-beam radiation therapy can be continuous hyperfractionated radiotherapy.
  • the external-beam radiation therapy can be intensity-modulated radiotherapy.
  • the external-beam radiation therapy can be helical tomotherapy.
  • the external-beam radiation therapy can be three dimensional conformal radiotherapy with dose escalation.
  • the external-beam radiation therapy can be stereotactic radiotherapy, including, but not limited to, single fraction stereotactic radiotherapy, fractionated stereotactic radiotherapy, and fractionated stereotactically guided conformal radiotherapy.
  • the external-beam radiation therapy can be generated or manipulated by any means known to one of skill in the art.
  • the photon beam used in external-beam radiation therapy can be shaped by a multileaf collimator.
  • suitable devices for generating a photon beam for use in external-beam radiation therapy include a gamma knife and a linac- based stereotactic apparatus.
  • administration of the external-beam radiation therapy is controlled by a computer according to a three-dimensional model of the patient in the treatment position.
  • Such a model can be generated, for example, by computed tomography (CT), magnetic resonance imaging (MRI), single photon emission computer tomography (SPECT), and positron emission tomography (PET).
  • CT computed tomography
  • MRI magnetic resonance imaging
  • SPECT single photon emission computer tomography
  • PET positron emission tomography
  • healthy tissues can optionally be protected from the effects of the external-beam radiation therapy by placing blocking devices such as, e.g., lead shields, in locations where such protection is needed.
  • blocking devices such as, e.g., lead shields
  • metal reflecting shields can optionally be located to reflect the photon beam in order to concentrate the radiation on the cancerous tissue to be treated and protect healthy tissue. Placement of either shield is well within the knowledge of one of skill in the art.
  • the present invention provides the administration of a vitamin D compound, or a mimic thereof, in combination with a treatment comprising a therapeutically effective dose of thermotherapy.
  • the thermotherapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation.
  • the thermotherapy can be cryoablation therapy.
  • the thermotherapy can be hyperthermic therapy.
  • the thermotherapy can be a therapy that elevates the temperature of the tumor higher than in hyperthermic therapy.
  • Cryoablation therapy involves freezing of a neoplastic mass, leading to deposition of intra- and extra-cellular ice crystals; disruption of cellular membranes, proteins, and organelles; and induction of a hyperosmotic environment, thereby causing cell death.
  • Cryoablation can be performed in one, two, or more freeze-thaw cycles, and further the periods of freezing and thawing can be adjusted for maximum tumor cell death by one of skill in the art.
  • One exemplary device that can be used in cryoablation is a cryoprobe incorporating vacuum-insulated liquid nitrogen. See, e.g., Murphy et al., Sent. Urol. Oncol. 79:133-140 (2001).
  • any device that can achieve a local temperature of about -18O 0 C to about -195 0 C can be used in cryoablation therapy.
  • Methods for and apparatuses useful in cryoablation therapy are described in U.S. Patent Nos. 6,383,181, 6,383,180, 5,993,444, 5,654,279, 5,437,673, and 5,147,355, each of which is incorporated herein by reference in its entirety.
  • Hyperthermic therapy typically involves elevating the temperature of a neoplastic mass to a range from about 42 0 C to about 44 0 C.
  • the temperature of the cancer may be further elevated above this range; however, such temperatures can increase injury to surrounding healthy tissue while not causing increased cell death within the tumor to be treated.
  • the tumor may be heated in hyperthermic therapy by any means known to one of skill in the art without limitation.
  • the tumor may be heated by microwaves, high intensity focused ultrasound, ferromagnetic thermoseeds, localized current fields, infrared radiation, wet or dry radiofrequency ablation, laser photocoagulation, laser interstitial thermic therapy, and electrocautery.
  • Microwaves and radiowaves can be generated by waveguide applicators, horn, spiral, current sheet, and compact applicators.
  • a vitamin D compound, or a mimic thereof is administered in combination with a treatment comprising a therapeutically effective dose of radiosurgery.
  • the radiosurgery can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation.
  • radiosurgery comprises exposing a defined volume within a subject to a manually directed radioactive source, thereby causing cell death within that volume.
  • the irradiated volume preferably contains the entire cancer to be treated, and preferably contains as little healthy tissue as possible.
  • the tissue to be treated is first exposed using conventional surgical techniques, then the radioactive source is manually directed to that area by a surgeon.
  • the radioactive source can be placed near the tissue to be irradiated using, for example, a laparoscope.
  • Methods and apparatuses useful for radiosurgery are further described in Valentini et al, Eur. J. Surg. Oncol. 28:180-185 (2002) and in U.S. Patent Nos. 6,421,416, 6,248,056, and 5,547,454, each of which is incorporated herein by reference in its entirety.
  • the vitamin D compound, or a mimic thereof is administered in combination with a treatment comprising a therapeutically effective dose of charged-particle radiotherapy.
  • the charged-particle radiotherapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation.
  • the charged-particle radiotherapy can be proton beam radiotherapy.
  • the charged-particle radiotherapy can be helium ion radiotherapy.
  • charged-particle radiotherapy comprises irradiating a defined volume within a subject with a charged-particle beam, thereby causing cellular death within that volume.
  • the irradiated volume preferably contains the entire cancer to be treated, and preferably contains as little healthy tissue as possible.
  • a method for administering charged-particle radiotherapy is described in U.S. Patent No. 5,668,371, which is incorporated herein by reference in its entirety.
  • the vitamin D compound, or a mimic thereof is administered in combination with a treatment comprising a therapeutically effective dose of neutron radiotherapy.
  • the neutron radiotherapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation.
  • the neutron radiotherapy can be a neutron capture therapy.
  • a compound that emits radiation when bombarded with neutrons and preferentially accumulates in a neoplastic mass is administered to a subject.
  • the tumor is irradiated with a low energy neutron beam, activating the compound and causing it to emit decay products that kill the cancerous cells.
  • Such compounds are typically boron containing compounds, but any compound that has a significantly larger neutron capture cross-section than common body constituents can be used.
  • the neutrons administered in such therapies are typically relatively low energy neutrons having energies at or below about 0.5 eV.
  • the compound to be activated can be caused to preferentially accumulate in the target tissue according to any of the methods useful for targeting of radionuclides, as described below, or in the methods described in Laramore, Semin. Oncol. 24:672-685 (1997) and in U.S. Patents Nos. 6,400,796, 5,877,165, 5,872,107, and 5,653,957, each of which is incorporated herein by reference in its entirety.
  • the neutron radiotherapy can be a fast neutron radiotherapy.
  • fast neutron radiotherapy comprises irradiating a defined volume within a subject with a neutron beam, thereby causing cellular death within that volume.
  • the irradiated volume preferably contains the entire cancer to be treated, and preferably contains as little healthy tissue as possible.
  • high energy neutrons are administered in such therapies, with energies in the range of about 10 to about 100 million eV.
  • fast neutron radiotherapy can be combined with charged-particle radiotherapy in the administration of mixed proton-neutron radiotherapy.
  • the vitamin D compound, or a mimic thereof is administered in combination with a treatment comprising a therapeutically effective dose of photodynamic therapy.
  • the photodynamic therapy can be administered according to any schedule, dose, or method known to one of skill in the art to be effective in the treatment or amelioration of cancer, without limitation, hi general, photodynamic therapy comprises administering a photosensitizing agent that preferentially accumulates in a neoplastic mass and sensitizes the neoplasm to light, then exposing the tumor to light of an appropriate wavelength. Upon such exposure, the photosensitizing agent catalyzes the production of a cytotoxic agent, such as, e.g., singlet oxygen, which kills the cancerous cells.
  • a cytotoxic agent such as, e.g., singlet oxygen
  • Representative photosensitizing agents that may be used in photodynamic therapy include, but are not limited to, porphyrins such as porfimer sodium, 5-aminolaevulanic acid and verteporfin; chlorins such as temoporfin; texaphyrins such as lutetium texephyrin; purpurins such as tin etiopurpurin; phthalocyanines; and titanium dioxide.
  • the wavelength of light used to activate the photosensitizing agent can be selected according to several factors, including the depth of the tumor beneath the skin and the absorption spectrum of the photosensitizing agent administered. The period of light exposure may also vary according to the efficiency of the absorption of light by the photosensitizing agent and the efficiency of the transfer of energy to the cytotoxic agent. Such determinations are well within the ordinary skill of one in the art.
  • vitamin D compounds can enhance the perfusion of cancerous tumors and, thereby, provide increased oxygen to the tumor thereby improving the sensitivity to radiotherapy.
  • the present invention enhances and expands the applicability of radiotherapy in the treatment or amelioration of cancer, that would otherwise not respond to current radiotherapy.
  • hyperproliferative disorders that ordinarily do not respond well to radiotherapy include, but are not limited to, oral melanoma, hemangiopericytomas, fibrosarcomas, and osteosarcomas.
  • sensitizing cells to treatment can allow use of a lower dose of radiotherapy, which reduces the side effects associated with the radiotherapy.
  • Radiotherapy can be administered to destroy tumor cells before or after surgery, before or after chemotherapy, and sometimes during chemotherapy. Radiotherapy may also be administered for palliative reasons to relieve symptoms of cancer, for example, to lessen pain.
  • Total body radiotherapy can be administered to patients who are undergoing a bone marrow transplant, which is a procedure often performed with subjects having leukemia, hi the case of a bone marrow transplant, a large single dose, or six to eight smaller doses of radiation, is administered to the whole body to destroy bone marrow cells in preparation for the transplant.
  • a bone marrow transplant which is a procedure often performed with subjects having leukemia, hi the case of a bone marrow transplant, a large single dose, or six to eight smaller doses of radiation, is administered to the whole body to destroy bone marrow cells in preparation for the transplant.
  • types of tumors that can be treated using radiotherapy are localized tumors that cannot be excised completely and metastases and tumors whose complete excision would cause unacceptable functional or cosmetic defects or be associated with unacceptable surgical risks
  • both the particular radiation dose to be utilized in treating cancer and the method of administration will depend on a variety of factors.
  • the dosages of radiation that can be used according to the methods of the present invention are determined by the particular requirements of each situation.
  • the dosage will depend on such factors as the size of the tumor, the location of the tumor, the age and sex of the patient, the frequency of the dosage, the presence of other tumors, possible metastases and the like.
  • Those skilled in the art of radiotherapy can readily ascertain the dosage and the method of administration for any particular tumor by reference to Hall, E. J., Radiobiology for the Radiobiologist, 5th edition, Lippincott Williams & Wilkins Publishers, Philadelphia, PA, 2000; Gunderson, L. L. and Tepper J.
  • vasodilators e.g., nitrates, calcium channel blockers
  • anticoagulants e.g., heparin
  • anti-platelet agents e.g., aspirin, blockers of Ilb/IIIa receptors, clopidogrel
  • anti -thrombins e.g., hirudin, iloprost
  • immunosuppressants e.g., sirolimus, tranilast, dexamethasone, tacrolimus, everolimus, A24
  • collagen synthetase inhibitors e.g., halofuginone, propyl hydroxylase, C-proteinase inhibitor, metalloproteinase inhibitor
  • antiinflammatories e.g., corticosteroids such as alclometasone, amcinonide, betamethasone, beclomethasone, budesonide, cortisone, clobeta
  • Anti-inflammatory drugs suitable for ameliorating tumor inflammation include salicylates (such as aspirin, choline magnesium trisalicylate, methyl salicylate, salsalte and diflunisal), acetic acids (such as indomethacin, sulindac, tolmetin, aceclofenac and diclofenac), 2-arylpropionic acids or profens (such as ibuprofen, ketoprofen, naproxen, fenoprofen, flurbiprofen and oxaprozin), N-arylanthranilic acids or fenamic acids (such as mefenamic acid, flufenamic acid, and meclofenamate), enolic acids or oxicams (such as piroxicam and meloxicam), cox inhibitors (such as celecoxib, rofecoxib (withdrawn from market), valdecoxib, parecoxib and etoricoxib), sulphonan
  • an immunomodulatory agent refers to an agent that modulates a host's immune system.
  • an immunomodulatory agent is an agent that alters the ability of a subject's immune system to respond to one or more foreign antigens.
  • an immunomodulatory agent is an agent that shifts one aspect of a subject's immune response, e.g., the agent shifts the immune response from a Th 1 to a Th2 response.
  • an immunomodulatory agent is an agent that inhibits or reduces a subject's immune system (i.e., an immunosuppressant agent). In certain other embodiments, an immunomodulatory agent is an agent that activates or increases a subject's immune system (i.e., an immunostimulatory agent).
  • Immunomodulatory agents useful for the present invention include, but are not limited to, small molecules, peptides, polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices and nucleotide sequences encoding biologically active proteins, polypeptides or peptides), antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules.
  • a particularly useful immunomodulatory agent useful for the present invention is thalidomide.
  • Immunosuppressant agents are useful to counteract autoimmune diseases, such as rheumatoid arthritis or Crohn's disease, and to prevent the immune system from attacking healthy parts of the body.
  • immunosuppressive agents useful for the present invention include glucocorticoid receptor agonists (e.g., cortisone, dexamethasone, hydrocortisone, betamethasone), calcineurin inhibitors (e.g., macrolides such as tacrolimus and pimecrolimus), immunophilins (e.g., cyclosporin A) and mTOR inhibitors (e.g., sirolimus, marketed as RAPAMUNE ® by Wyeth).
  • glucocorticoid receptor agonists e.g., cortisone, dexamethasone, hydrocortisone, betamethasone
  • calcineurin inhibitors e.g., macrolides such as tacrolimus and pimecrolimus
  • immunomodulatory agents useful for the present invention further include antiproliferative agents (e.g., methotrexate, leflunomide, cisplatin, ifosfamide, paclitaxel, taxanes, topoisomerase I inhibitors (e.g., CPT-I l, topotecan, 9- AC, and GG-211), gemcitabine, vinorelbine, oxaliplatin, 5-fluorouracil (5-FU), leucovorin, vinorelbine, temodal, taxol, cytochalasin B, gramicidin D, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, melphalan, glucocor
  • a vitamin D compound, or a mimic thereof, in combination with one or more therapeutic agents is intended to refer to the combined administration of a vitamin D compound, or a mimic thereof, and one or more therapeutic agents or treatments, wherein the vitamin D compound, or the mimic thereof, can be administered prior to, concurrently with, or after the administration of the therapeutic agents or treatments.
  • the vitamin D compound, or the mimic thereof can be administered up to two weeks or more prior to or after the therapeutic agents or treatments and still be considered to be a combination treatment.
  • vitamin D compound is intended to refer to a vitamin D compound that is or becomes biologically active when administered to a subject or contacted with cells.
  • biological activity refers to any known in vivo activity of vitamin D, e.g., binding to and stimulating the vitamin D receptor.
  • Active vitamin D compounds include compounds that cause hypercalcemia and compounds that do not cause hypercalcemia upon administration.
  • the biological activity of a vitamin D compound can be assessed by assays well known to one of skill in the art such as, e.g., immunoassays that measure the expression of a specific gene regulated by vitamin D. Vitamin D compounds exist in several forms with different levels of activity in the body.
  • a vitamin D compound may be partially activated by first undergoing hydroxylation in the liver at the carbon-25 position and then may be fully activated in the kidney by further hydroxylation at the carbon- 1 position.
  • the prototypical active vitamin D compound is l ⁇ ,25-hydroxyvitamin D 3 , also known as calcitriol.
  • the vitamin D compound may also be a partially hydroxylated vitamin D such as l ⁇ -hydroxyvitamin D3, also known as l ⁇ -calcidol, and 25-hydroxyvitamin D 3 also known as calcifediol.
  • a large number of other vitamin D compounds are known and can be used in the practice of the invention.
  • the vitamin D compounds include, but are not limited to, analogs, homologs, mimics, and derivatives of vitamin D compounds such as those described in the following patents: U.S. Patent Nos. 4,391,802 (l ⁇ -hydroxyvitamin D derivatives); 4,717,721 (l ⁇ -hydroxy derivatives with a 17 side chain greater in length than the cholesterol or ergosterol side chains); 4,851,401 (cyclopentano-vitamin D analogs); 4,866,048 and 5,145,846 (vitamin D 3 analogues with alkynyl, alkenyl, and alkanyl side chains); 5,120,722 (trihydroxycalciferol); 5,547,947 (fluoro-cholecalciferol compounds); 5,446,035 (methyl substituted vitamin D); 5,411,949 (23-oxa-derivatives); 5,237,110 (19-nor- vitamin D compounds; 4,857,518 (hydroxylated 24-homo-vitamin D derivatives).
  • ROCALTROL Roche Laboratories
  • CALCIJEX injectable calcitriol investigational drugs from Leo Pharmaceuticals including EB 1089 (24a,26a,27a-trihomo-22,24-diene-l ⁇ ,25-(OH) 2 -D 3 , KH 1060 (20-epi-22-oxa- 24a,26a,27a-trihomo-l ⁇ ,25-(OH) 2 -D 3 ), MC 1288 (l,25-(OH) 2 -20-epi-D 3 ) and MC 903 (calcipotriol, l ⁇ 24s-(OH) 2 -22-ene-26,27-dehydro-D 3 ); Roche Pharmaceutical drugs that include 1, 25-(OH) 2 - 16-ene-D 3 , 1, 25-(OH) 2 - 16-ene- 23-yne-D 3 , and 25-(OH) 2 - 16-ene-23-yne-D 3 ; Chugai Pharmaceutical
  • Additional examples include l ⁇ ,25-(OH) 2 - 26,27-d 6 -D 3 ; l ⁇ ,25-(OH) 2 -22-ene-D 3 ; l ⁇ ,25-(OH) 2 -D 3, l ⁇ ,25-(OH) 2 -D 2 ; l ⁇ ,25-(OH) 2 -D 4 ; l ⁇ ,24,25-(OH) 3 -D 3 ; l ⁇ ,24,25-(OH) 3 -D 2 ; l ⁇ ,24,25-(OH) 3 -D 4 ; l ⁇ -(OH)-25-FD 3 ; l ⁇ -(OH)-25-FD 4 ; l ⁇ -(OH)-25-FD 2 ; l ⁇ ,24-(OH) 2 -D 4 ; l ⁇ ,24- (OH) 2 -D 3 ; l ⁇ ,24-(OH) 2 -D 3 ; l ⁇ ,24- (OH) 2 -D 3 ; l ⁇ ,
  • U.S. Patent No. 6,521,608 See also, e.g., U.S. Patent Nos. 6,689,766, 6,503,893, 6,482,812, 6,441,207, 6,410,523, 6,399,797, 6,392,071, 6,376,480, 6,372,926, 6,372,731, 6,359,152, 6,329,357, 6,326,503, 6,310,226, 6,288,249, 6,281,249, 6,277,837, 6,218,430, 6,207,656, 6,197,982, 6,127,559, 6,103,709, 6,080,878, 6,075,015, 6,072,062, 6,043,385, 6,017,908, 6,017,907,
  • the term "mimic” as used herein is intended to refer to non- secosteroidal vitamin D mimic compounds.
  • these non- secosteroidal vitamin D mimics are compounds that do not structurally fall within the class of compounds generally known as vitamin D compounds but which modulate the activity of vitamin D nuclear receptors.
  • Examples of such vitamin D mimics include bis-aryl derivatives disclosed by U.S. Patent 6,218,430 and WO publication 2005/037755. Additional examples of non-secosteroidal vitamin D mimic compounds suitable for the present invention can be found in U.S.
  • the invention is drawn to methods employing non-secosteroidal vitamin D mimic compounds having Formula I:
  • R 1 and R 2 are each independently halo, haloalkyl, pseudohalo, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl or optionally substituted heteroaryl; or
  • R 1 and R 2 together with the carbon atom to which they are attached, form an optionally substituted cycloalkyl consisting of:
  • k is an integer from 1 to 6;
  • R 1 and R 2 together with the carbon atom to which they are attached, form an optionally substituted heterocyclyl selected from a group consisting of:
  • A is -O-, -NR X -, -S-, -S(O)- or -S(O) 2 - wherein R x is hydrogen, alkyl, haloalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -R 14 -C(J)R 15 , -R 14 -C(J)OR 15 , -R 14 -C(J)R 16 OR 15 , -R 14 -C(J)SR 16 , -R 14 -C(J)N(R 18 )R 19 , -R 14 -C(J)N(R 17 )N(R 18 )R 19 , -R 14 -C(J)N(R I7 )S(O) p R 20 , -R 14 -S(O) P N(R 18 )R 19 , or -R 14 -S(O) p R 20 ; and wherein B is -O-
  • R 3 and R 4 are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, pseudohalo, nitro, cyano, azido, -R 14 -OR 15 , -R 14 -N(R 18 )R 19 , -R I4 -SR 15 , -R 14 -OC(J)R 15 , -R 14 -NR 17 C(J)R 15 , -R 14 -OC(J)N(R 18 )R 19 , -R 14 -NR 17 C(J)N(R 18 )R 19 ,
  • R 5 , R 6 , R 7 , R 8 , R 9 , R 10 are each independently hydrogen, halo, hydroxy, amino, pseudohalo, cyano, nitro, alkyl, haloalkyl, alkoxy or haloalkoxy;
  • X is R 25 ;
  • Y is independently R 30 , -OR 31 , -SR 32 or -N(R 33 )(R 34 );
  • R 25 and R 30 are each independently selected from (i) or (ii) as follows:
  • optionally substituted alkyl that may be substituted with one to ten substituents each independently selected from a group consisting of halo, pseudohalo, nitro, cyano, thioxo, azido, amidino, guanidino, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heteroaryl, optionally substituted heteroaralkyl, -OR 15 , -OR 16 OR 15 , -N(R 18 )R 19 , -N(R 17 )N(R 18 )R 19 , -SR 15 , -SR 16 SR 15 , -N(R 17 )N(R 17 )S(O) P R 20 , -OC(J)R 15 , -NR 17 C(J)R 15 ,
  • optionally substituted alkenyl or optionally substituted alkynyl may be substituted with one to ten substituents each independently selected from a group consisting of oxo, thioxo, halo, pseudohalo, nitro, cyano, azido, amidino, guanidino, -OR 15 , -OR 16 OR 15 , -N(R 18 )R 19 , -N(R 17 )N(R 18 )R 19 , -SR 15 , -SR 16 SR 15 , -S(O) P R 20 , -N(R 17 )S(O) p R 20 , -N(R 17 )N(R 17 )S(O) p R 20 , -OC(J)R 15 , -NR 17 C(J)R 15 , -OC(J)N(R 18 )R 19 , -NR 17 C(J)N(R 18 )R 19 , -NR 17 C(
  • R 31 ' R 32 , R 33 , and R 34 are each independently optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl or optionally substituted cycloalkyl; all of which may be optionally substituted with one to ten substituents each independently selected from a group consisting of oxo, halo, pseudohalo, nitro cyano, azido, amidino, guanidino -OR 15 , -OR 16 OR 15 , -N(R 18 )R 19 , -N(R 17 )N(R 18 )R 19 , -SR 15 , -SR 16 SR 15 , -S(O)pR 20 , -N(R 17 )S(O)pR 20 , -N(R 17 )N(R 17 )S(O) p R 20 , -OC(J)R 15 , -NR 17 C(J)R 15 , -OC(J)
  • R 34 can additionally be hydrogen; where each R 14 is independently a direct bond or alkylene; where each R 15 and R 17 is independently hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted aryl or optionally substituted heteroaryl, all of which, when substituted, are substituted with one to five substituents each independently selected from halo, cyano, hydroxy and amino; where each R 16 and R 20 is independently optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted
  • R 22 is hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl or aralkyl;
  • R 23 and R 24 are each independently hydrogen, alkyl, haloalkyl, alkenyl, alkynyl or cycloalkyl; or R 23 and R 24 , together with the nitrogen atom to which they are attached, form a heterocyclyl or heteroaryl; each J is independently O or S; as a single isomer, a mixture of isomers, or as a racemic mixture of isomers; as a solvate or polymorph; or as a prodrug or metabolite; or as a pharmaceutically acceptable salt thereof.
  • R 1 and R 2 may form a substituted cyclohexyl, said cyclohexyl, when substituted at the 4-position relative to the gem-diaryl substituents, may be substituted with a substituent selected from the group consisting of halo, cyano, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R 25 and R 30 are not -CH 2 COOH;
  • E and F are each independently selected from the group consisting of O, S, and NR 41 ;
  • R 35 and R 36 are independently selected from the group consisting of alkyl groups, optionally fluorinated; or together R 35 and R 36 form a cycloalkylidene having 3 to 8 carbon atoms, optionally fluorinated;
  • R and R are independently selected from the group consisting of halogen; lower n-alkyl, optionally fluorinated; and lower alkoxy, optionally fluorinated;
  • R 39 is selected from the group consisting of H; optionally substituted alkyl groups; optionally substituted alkenyl groups; optionally substituted alkynyl groups; optionally substituted aryl groups; OR 45 ; NR 46 R 47 ; or together with R 42 , R 43 , or R 44 forms a 3- to 12-membered cyclic group wherein said cyclic group is selected from the group consisting of amidines, amines, ethers, lactams, lactones, ketals, hemiketals, aminals, hemiaminals, carbonates, carbamates, ureas, and combinations thereof; R 40 is selected from the group consisting of H and alkyl groups, optionally substituted;
  • R 41 is selected from the group consisting of H and alkyl groups, optionally substituted;
  • R 42 is selected from the group consisting of H, optionally substituted alkyl groups, optionally substituted alkenyl groups, optionally substituted alkynyl groups, optionally substituted aryl group, and optionally substituted acyl groups;
  • R 43 and R 44 are independently selected from the group consisting of H, optionally substituted alkyl groups, optionally substituted alkenyl groups, optionally substituted alkynyl groups, optionally substituted aryl groups, and optionally substituted acyl groups;
  • R 45 is selected from the group consisting of H, optionally substituted alkyl groups, optionally substituted alkenyl groups, optionally substituted alkynyl groups, optionally substituted aryl groups, and optionally substituted acyl groups;
  • R 46 and R 47 are independently selected from the group consisting of H, optionally substituted alkyl groups, optionally substituted alkenyl groups, optionally substituted alkynyl groups, optionally substituted aryl groups, and optionally substituted acyl groups and pharmaceutically acceptable salts thereof.
  • R 45 is selected from the group consisting of OH and Ci -C 4 alkoxy
  • R 46 is not carboxymethyl and alkyl esters thereof.
  • R 45 is not H or primary alkyl.
  • R 46 and R 48 do not both comprise aziridines.
  • R 45 , R 46 , and R 48 do not simultaneously comprise alkenyl ethers.
  • R 45 and R 46 do not both comprise glycidyl ethers.
  • HDPA high dose pulse administration
  • an inhibitor of the cytochrome p450 enzyme 24-hydroxylase (also known as CYP24) can be administered instead of or in addition to the vitamin D compound or a mimic thereof.
  • 24-hydroxylase which is the primary enzyme involved in the metabolism of vitamin D
  • the increased vitamin D level itself may lead to a therapeutic effect or allow the administration of lower doses of a vitamin D compound or a mimic thereof to achieve the same therapeutic effect.
  • Any 24-hydroxylase inhibitor known in the art may be used, including agents that inhibit 24- hydroxyalse activity by at least about 10, 20, 30, 40, 50, 60, 70, 80, or 90%.
  • any agent to inhibit 24-hydroxylase activity may be measured using routine techniques known in the art, such as those disclosed in U.S. Published Application No. 2006/0078494.
  • 24-hydroxylase inhibitors include, without limitation, azoles such as the antifungals ketoconazole, clotrimazole, itraconazole, and fluconazole, compounds such as (R)-SDZ-286907, (R)-SDZ-287871, (R)-VAB636, (S)-SDZ-285428, (R)- VID400, and the agents disclosed in U.S. Published Application No. 2006/0078494, US Published Application No. 2006/0074109 and Kahraman et al, J. Med. Chem. 47:6854 (2004), each incorporated be reference herein.
  • hypocalcemia refers to a medical condition in which the concentration of calcium ions in the plasma is greater than about 10.5 mg/dL in humans.
  • symptomatic hypercalcemia refers to symptoms associated with one of more of the signs or symptoms of hypercalcemia.
  • Early manifestations of hypercalcemia include weakness, headache, somnolence, nausea, vomiting, dry mouth, constipation, muscle pain, bone pain, or metallic taste.
  • Late manifestations include polydypsia, polyuria, weight loss, pancreatitis, photophobia, pruritis, renal dysfunction, aminotransferase elevation, hypertension, cardiac arrhythmias, psychosis, stupor, or coma.
  • Methods to determine the concentration of calcium ions in blood plasma are generally within the capability of a person of ordinary skill in the art.
  • grade 4 toxicity is associated with reduced count for WBC, platelets, hemoglobin, neutrophils and lymphocytes; massive hemorrhage; gastrointestinal problems (such as vomiting more than 10 times a day, diarrhea (>10 times a day) and stomatitis which requires IV nutrition); hepatic failures (such as elevated bilirubin and hepatic coma), kidney/bladder dysfunction; cardiovascular events (such as refractory congestive heart failure, acute myocardial infraction, dyspnea at rest and cardiac tamponade); neuralgic disorders (such as paralysis, coma, seizures, cerebellar necrosis, severe headaches, blindness, uncorrectable deafness and suicidal mood) and metabolic problems (such as hyperg
  • grade 3 toxicity is milder than grade 4 toxicity, it can be life threatening and is associated with reduced count for WBC, platelets, hemoglobin, neutrophils and lymphocytes; gross hemorrhage; gastrointestinal problems (such as vomiting 6-10 times a day, diarrhea (7-9 times a day) and painful ulcers (patient could not eat)); hepatic failures (such as precoma and elevated bilirubin); cardiovascular events (such as mild congestive heart failure responsive to treatment, angina without infraction and symptomatic effusion); neurologic disorders (such as severe loss or impairment of neurosensory, severe cortical contusion, unrelenting headache and correctable hearing loss) and weight change.
  • the vitamin D compound or mimic thereof has a reduced hypercalcemic effect as compared to vitamin D so that sufficient doses of the compound can be administered without inducing hypercalcemia in the animal.
  • the reduced hypercalcemia effect may be due to the active vitamin D compound itself, the regimen by which the compound is administered, or both.
  • a reduced hypercalcemic effect is defined as an effect which is less than the hypercalcemic effect induced by administration of an equal dose of l ⁇ ,25-hydroxyvitamin D 3 (calcitriol).
  • EB 1089 has a hypercalcemic effect which is 50% of the hypercalcemic effect of calcitriol.
  • Additional vitamin D compounds having a reduced hypercalcemic effect include Ro23-7553 and Ro24-5531 available from Hoffmann LaRoche.
  • Other examples of vitamin D compounds having a reduced hypercalcemic effect can be found in U.S. Patent No. 4,717,721. Determining the hypercalcemic effect of an vitamin D compound is routine in the art and can be carried out as disclosed in Hansen et al, Curr. Pharm. Des. 6: 803-828 (2000).
  • a vitamin D compound is administered to an animal before, during and/or after chemotherapy.
  • the vitamin D compound can be administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week,
  • the vitamin D compound can be administered 1 hour, 2 hours,
  • the vitamin D compound is administered before, during, and after the chemotherapy or radiotherapy.
  • one or more therapeutic agents or treatments are administered to an animal in addition to the vitamin D compound.
  • the vitamin D compound, or a mimic thereof can be administered prior to ⁇ e.g., 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 36 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 2 weeks, 3 weeks, 4 weeks or more), concurrently with, or after ⁇ e.g., 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 36 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 2 weeks, 3 weeks, 4 weeks or more) the administration of one or more therapeutic agents or treatments.
  • the method of administering a vitamin D compound, or a mimic thereof, in combination with one or more therapeutic agents or treatments may be repeated at least once.
  • the method may be repeated as many times as necessary to achieve or maintain a therapeutic response, e.g., from one to about ten times.
  • the vitamin D compound, or a mimic thereof, and the one or more therapeutic agents or treatments may be the same or different from that used in the previous repetition.
  • the time period of administration of the vitamin D compound, or a mimic thereof, and the manner in which it is administered can vary from repetition to repetition.
  • the doses of the vitamin D compounds and mimics may be adjusted proportionate to the ratio of the efficacy index to the calcemic index according to the formula:
  • Dose CalcitriolDose x (E/ ⁇ CI)
  • Dose is the vitamin D compound or mimic dose
  • calcitriolDose is calcitriol dose
  • EI is the analog or mimic efficacy index
  • CI is the vitamin D compound or mimic calcemic index
  • the term "efficacy index” is the ratio of the concentration of the vitamin D compound or mimic to the concentration of calcitriol at equivalent potency.
  • the efficacy index is a fraction less than one when the vitamin D compound or mimic is less potent than calcitriol.
  • EI is number greater than one when calcitriol is less potent than the vitamin D compound or mimic.
  • the "calcemic index" of a drug is a measure of the relative ability of the drug to generate a calcemic response as reported in Bouillon et al, Endocrine Reviews 16:200-251, 1995.
  • a calcemic index of 1 corresponds to the relative calcemic activity of calcitriol.
  • a calcemic index of about 0.01 corresponds to the calcemic activity of a drug with approximately 100 times less calcemic activity than calcitriol.
  • a calcemic index of 0.5 would correspond to a drug having approximately half the calcemic activity of calcitriol.
  • the calcemic index of a drug can vary depending on the assay conducted, e.g., whether one is measuring stimulation of intestinal calcium absorption (a process by which dietary calcium enters into the physiological processes to contribute to the skeletal growth of the organism and to the maintenance of calcium homeostasis) or bone calcium mobilizing activity (a process by which the bone matrix acts as an exchangeable reservoir for calcium). See U.S. Patent 6,521,608 for further detail.
  • the vitamin D compound or a mimic thereof is preferably administered at a dose of about 0.1 ⁇ g to about 10 mg, e.g., about 0.5 ⁇ g to about 1 mg, or from about 15 ⁇ g to about 500 ⁇ g.
  • an effective amount of an vitamin D compound or a mimic thereof is 0.5, 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, or 10000 ⁇ g or more.
  • an effective dose of a vitamin D compound or a mimic thereof is between about 3 ⁇ g to about 10 mg, e.g., between about 15 ⁇ g to about 1 mg, between about 30 ⁇ g to about 300 ⁇ g, between about 35 ⁇ g to about 200 ⁇ g, between about 40 ⁇ g to about 100 ⁇ g, or about 45 ⁇ g.
  • an effective amount of a vitamin D compound or a mimic thereof is about 300, 400, 500, 600, 700, 800, 900 ⁇ g, 1, 2, 3, 4 or 5 mg.
  • an effective dose of a vitamin D compound or a mimic thereof is between about 300 ⁇ g to about 5 mg, more preferably between about 500 ⁇ g and about 4 mg, more preferably between about 800 ⁇ g and about 3 mg, more preferably between about 1 and about 3 mg.
  • the methods of the invention comprise administering a vitamin D compound or a mimic thereof in a dose of about 0.12 ⁇ g/kg bodyweight to about 200 ⁇ g/kg bodyweight.
  • the compound may be administered by any route, including oral, intramuscular, intravenous, subcutaneous, parenteral, rectal, nasal, topical, or transdermal.
  • the dose may be kept low, for example about 0.5 ⁇ g to about 5 ⁇ g, in order to avoid or diminish the induction of hypercalcemia. If the vitamin D compound or a mimic thereof has a reduced hypercalcemic effect a higher daily dose may be administered without resulting in hypercalcemia, for example about 10 ⁇ g to about 20 ⁇ g or higher (up to about 50 ⁇ g to about 100 ⁇ g)-
  • the vitamin D compound or a mimic thereof is administered by HDPA so that high doses of the vitamin D compound or the mimic thereof can be administered without inducing hypercalcemia.
  • HDPA refers to intermittently administering a vitamin D compound, or a mimic thereof, on either a continuous intermittent dosing schedule or a non-continuous intermittent dosing schedule.
  • High doses of vitamin D compounds include doses greater than about 3 ⁇ g as discussed in the sections above.
  • the frequency of the HDPA can be limited by a number of factors including, but not limited to, the pharmacokinetic parameters of the compound or formulation and the pharmacodynamic effects of the vitamin D compound, or a mimic thereof, on the animal. For example, animals having impaired renal function may require less frequent administration of the vitamin D compound or the mimic thereof because of the decreased ability of those animals to excrete calcium.
  • HDPA can encompass any discontinuous administration regimen designed by a person of skill in the art.
  • the vitamin D compound, or the mimic thereof can be administered not more than once every three days, every four days, every five days, every six days, every seven days, every eight days, every nine days, every ten days, every two weeks, every three weeks, or every four weeks.
  • the administration can continue for one, two, three, or four weeks or one, two, or three months, or longer.
  • the vitamin D compound, or the mimic thereof can be administered under the same or a different schedule.
  • the period of rest can be one, two, three, or four weeks, or longer, according to the pharmacodynamic effects of the vitamin D compound, or the mimic thereof, on the animal.
  • the vitamin D compound or the mimic thereof can be administered once per week for three months.
  • the vitamin D compound or a mimic thereof can be administered once per week for three weeks of a four week cycle. After a one week period of rest, the vitamin D compound or a mimic thereof can be administered under the same or different schedule.
  • an effective dose of a vitamin D compound, or a mimic thereof is any dose of the compound effective to prevent, treat, or ameliorate a hyperproliferative disease.
  • a high dose of a vitamin D compound, or a mimic thereof can be a dose from about 3 ⁇ g to about 10 mg or any dose within this range as discussed above.
  • the dose, dose frequency, duration, or any combination thereof may also vary according to age, body weight, response, and the past medical history of the animal as well as the route of administration, pharmacokinetics, and pharmacodynamic effects of the pharmaceutical agents or treatments. These factors are routinely considered by one of skill in the art.
  • the rate of absorption and clearance of vitamin D compounds and mimics thereof are affected by a variety of factors that are well known to persons of skill in the art. As discussed above, the pharmacokinetic properties of vitamin D compounds and mimics thereof limit the peak concentration of vitamin D compounds and mimics thereof that can be obtained in the blood without inducing the onset of hypercalcemia. The rate and extent of absorption, distribution, binding or localization in tissues, biotransformation, and excretion of the vitamin D compound or a mimic thereof can all affect the frequency at which the pharmaceutical agents or treatments can be administered.
  • a vitamin D compound or a mimic thereof is administered at a dose sufficient to achieve peak plasma concentrations of the vitamin D compound, or the mimic thereof, of about 0.1 nM to about 1000 nM, e.g., about 0.1 nM to about 25 nM.
  • the methods of the invention comprise administering the vitamin D compound, or the mimic thereof, in a dose that achieves peak plasma concentrations of 0.1 nM, 0.2 nM, 0.3 nM, 0.4 nM, 0.5 nM, 0.6 nM, 0.7 nM, 0.8 nM, 0.9 nM, 1 nM, 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 12.5 nM, 15 nM, 17.5 nM, 20 nM, 22.5 nM, 25 nM, 30 nM, 35 nM, 40 nM, 45 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 150 nM, 200 nM, 250 nM, 300 nM, 350 nM, 400 nM, 450
  • the vitamin D compound, or the mimic thereof is administered in a dose that achieves peak plasma concentrations of the vitamin D compound, or the mimic thereof, exceeding about 0.5 nM, e.g., about 0.5 nM to about 1000 nM, about 0.5 nM to about 100 nM, about 0.5 nM to about 25 nM, about 5 nM to about 20 nM, or about 10 nM to about 15 nM.
  • 0.5 nM e.g., about 0.5 nM to about 1000 nM, about 0.5 nM to about 100 nM, about 0.5 nM to about 25 nM, about 5 nM to about 20 nM, or about 10 nM to about 15 nM.
  • the vitamin D compound, or a mimic thereof is administered at a dose of at least about 0.12 ⁇ g/kg bodyweight, more preferably at a dose of at least about 0.5 ⁇ g/kg bodyweight.
  • the methods of the invention further comprise administering a dose of a vitamin D compound, or a mimic thereof, that achieves peak plasma concentrations rapidly, e.g., within four hours.
  • the methods of the invention comprise administering a dose of a vitamin D compound, or a mimic thereof, that is eliminated quickly, e.g., with an elimination half-life of less than 12 hours.
  • the methods of the invention encompass HDPA of vitamin D compounds, or mimics thereof, to an animal before, during, or after chemotherapy or radiotherapy and monitoring the animal for symptoms associated with hypercalcemia.
  • symptoms include calcification of soft tissues (e.g., cardiac tissue), increased bone density, and hypercalcemic nephropathy.
  • the methods of the invention encompass HDPA of a vitamin D compound, or a mimic thereof, to an animal before, during, or after chemotherapy or radiotherapy and monitoring the calcium plasma concentration of the animal to ensure that the calcium plasma concentration is less than about 11.5 mg/dL.
  • high blood levels of vitamin D compounds can be safely obtained in conjunction with reducing the transport of calcium into the blood.
  • higher vitamin D compound concentrations are safely obtainable without the onset of hypercalcemia when administered in conjunction with a reduced calcium diet.
  • the calcium can be trapped by an adsorbent, absorbent, ligand, chelate, or other binding moiety that cannot be transported into the blood through the small intestine.
  • the rate of osteoclast activation can be inhibited by administering, for example, a bisphosphonate such as, e.g., zoledronate, pamidronate, or alendronate, or a corticosteroid such as, e.g., dexamethasone or prednisone, in conjunction with the vitamin D compound, or a mimic thereof.
  • a bisphosphonate such as, e.g., zoledronate, pamidronate, or alendronate
  • a corticosteroid such as, e.g., dexamethasone or prednisone
  • high blood levels of vitamin D compounds are safely obtained in conjunction with maximizing the rate of clearance of calcium.
  • calcium excretion can be increased by ensuring adequate hydration and salt intake.
  • diuretic therapy can be used to increase calcium excretion.
  • the vitamin D compound or a mimic thereof may be administered as part of a pharmaceutical composition comprising a pharmaceutically acceptable carrier, wherein the vitamin D compound or a mimic thereof is present in an amount which is effective to achieve its intended purpose, i.e., to have the desired effect of preventing, treating or ameliorating a disorder in the animal receiving chemotherapy or radiotherapy.
  • the pharmaceutical composition may further comprise one or more excipients, diluents or any other components known to persons of skill in the art and germane to the methods of formulation of the present invention.
  • the pharmaceutical composition may additionally comprise other compounds typically used as adjuncts during chemotherapy.
  • composition as used herein is to be understood as defining compositions of which the individual components or ingredients are themselves pharmaceutically acceptable, e.g., where oral administration is foreseen, acceptable for oral use and, where topical administration is foreseen, topically acceptable.
  • the pharmaceutical composition can be prepared in single unit dosage forms.
  • the dosage forms are suitable for oral, mucosal (nasal, sublingual, vaginal, buccal, rectal), parenteral (intravenous, intramuscular, intraarterial), or topical administration.
  • Preferred dosage forms of the present invention include oral dosage forms and intravenous dosage forms.
  • Intravenous forms include, but are not limited to, bolus and drip injections.
  • the intravenous dosage forms are sterile or capable of being sterilized prior to administration to a subject since they typically bypass the subject's natural defenses against contaminants.
  • intravenous dosage forms include, but are not limited to, Water for Injection USP; aqueous vehicles including, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles including, but not limited to, ethyl alcohol, polyethylene glycol and polypropylene glycol; and non-aqueous vehicles including, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate and benzyl benzoate.
  • aqueous vehicles including, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles including, but not limited to, ethyl alcohol, polyethylene glycol and polyprop
  • the pharmaceutical compositions comprising vitamin D compounds, or a mimic thereof are emulsion pre-concentrate formulations.
  • the compositions of the invention meet or substantially reduce the difficulties associated with vitamin D compound therapy or a mimic thereof hitherto encountered in the art including, in particular, undesirable pharmacokinetic parameters of the compound upon administration to the animal.
  • a pharmaceutical composition comprising (a) a lipophilic phase component, (b) one or more surfactants, (c) a vitamin D compound, or a mimic thereof; wherein said composition is an emulsion pre-concentrate, which upon dilution with water, in a water to composition ratio of about 1:1 or more of said water, forms an emulsion having an absorbance of greater than 0.3 at 400 nm.
  • the pharmaceutical composition of the invention may further comprise a hydrophilic phase component.
  • a pharmaceutical emulsion composition comprising water (or other aqueous solution) and an emulsion pre-concentrate.
  • emulsion pre-concentrate is intended to mean a system capable of providing an emulsion upon contacting with, e.g., water.
  • emulsion as used herein, is intended to mean a colloidal dispersion comprising water and organic components including hydrophobic (lipophilic) organic components.
  • emulsion is intended to encompass both conventional emulsions, as understood by those skilled in the art, as well as “sub-micron droplet emulsions,” as defined immediately below.
  • sub-micron droplet emulsion as used herein is intended to mean a dispersion comprising water and organic components including hydrophobic (lipophilic) organic components, wherein the droplets or particles formed from the organic components have an average maximum dimension of less than about 1000 nm.
  • Sub-micron droplet emulsions are identifiable as possessing one or more of the following characteristics. They are formed spontaneously or substantially spontaneously when their components are brought into contact, that is without substantial energy supply, e.g., in the absence of heating or the use of high shear equipment or other substantial agitation. They exhibit thermodynamic stability and they are monophasic.
  • the particles of a sub-micron droplet emulsion may be spherical, though other structures are feasible, e.g. liquid crystals with lamellar, hexagonal or isotropic symmetries.
  • sub-micron droplet emulsions comprise droplets or particles having a maximum dimension ⁇ e.g., average diameter) of between about 50 nm to about 1000 nm, and preferably between about 200 nm to about 300 nm.
  • the pharmaceutical compositions of the present invention will generally form an emulsion upon dilution with water.
  • the emulsion will form according to the present invention upon the dilution of an emulsion pre- concentrate with water in a water to composition ratio of about 1 : 1 or more of said water.
  • the ratio of water to composition can be, e.g., between 1 :1 and 5000:1.
  • the ratio of water to composition can be about 1:1, 2:1, 3:1, 4:1, 5:1, 10:1, 200:1, 300:1, 500:1, 1000:1, or 5000:1.
  • the skilled artisan will be able to readily ascertain the particular ratio of water to composition that is appropriate for any given situation or circumstance.
  • an emulsion upon dilution of said emulsion pre- concentrate with water, an emulsion will form having an absorbance of greater than 0.3 at 400 nm.
  • the absorbance at 400 nm of the emulsions formed upon 1 : 100 dilution of the emulsion pre-concentrates of the present invention can be, e.g., between 0.3 and 4.0.
  • the absorbance at 400 nm can be about 0.4, 0.5, 0.6, 1.0, 1.2, 1.6, 2.0, 2.2, 2.4, 2.5, 3.0, or 4.0.
  • Methods for determining the absorbance of a liquid solution are well known by those in the art.
  • compositions of the present invention can be, e.g., in a solid, semi-solid, or liquid formulation.
  • Semi-solid formulations of the present invention can be any semi-solid formulation known by those of ordinary skill in the art, including, e.g., gels, pastes, creams and ointments.
  • compositions of the present invention comprise a lipophilic phase component.
  • Suitable components for use as lipophilic phase components include any pharmaceutically acceptable solvent which is non- miscible with water. Such solvents will appropriately be devoid or substantially devoid of surfactant function.
  • the lipophilic phase component may comprise mono-, di- or triglycerides.
  • Mono-, di- and triglycerides that may be used within the scope of the invention include those that are derived from C 6 , C 8 , C 10 , C 12 , C 14 , C 16 , Cj 8 , C 20 and C 22 fatty acids.
  • Exemplary diglycerides include, in particular, diolein, dipalmitolein, and mixed caprylin-caprin diglycerides.
  • Preferred triglycerides include vegetable oils, fish oils, animal fats, hydrogenated vegetable oils, partially hydrogenated vegetable oils, synthetic triglycerides, modified triglycerides, fractionated triglycerides, medium and long-chain triglycerides, structured triglycerides, and mixtures thereof.
  • preferred triglycerides include: almond oil; babassu oil; borage oil; blackcurrant seed oil; canola oil; castor oil; coconut oil; corn oil; cottonseed oil; evening primrose oil; grapeseed oil; groundnut oil; mustard seed oil; olive oil; palm oil; palm kernel oil; peanut oil; rapeseed oil; safflower oil; sesame oil; shark liver oil; soybean oil; sunflower oil; hydrogenated castor oil; hydrogenated coconut oil; hydrogenated palm oil; hydrogenated soybean oil; hydrogenated vegetable oil; hydrogenated cottonseed and castor oil; partially hydrogenated soybean oil; partially soy and cottonseed oil; glyceryl tricaproate; glyceryl tricaprylate; glyceryl tricaprate; glyceryl triundecanoate; glyceryl trilaurate; glyceryl trioleate; glyceryl trilinoleate; glyceryl tri
  • the triglyceride is the medium chain triglyceride available under the trade name LABRAFAC CC.
  • Other preferred triglycerides include neutral oils, e.g., neutral plant oils, in particular fractionated coconut oils such as known and commercially available under the trade name MIGLYOL, including the products: MIGLYOL 810; MIGLYOL 812; MIGLYOL 818; and CAPTEX 355.
  • caprylic-capric acid triglycerides such as known and commercially available under the trade name MYRITOL, including the product MYRITOL 813.
  • Further suitable products of this class are CAPMUL MCT, CAPTEX 200, CAPTEX 300, CAPTEX 800, NEOBEE M5 and MAZOL 1400.
  • the lipophilic phase component is the product
  • compositions of the present invention may further comprise a hydrophilic phase component.
  • the hydrophilic phase component may comprise, e.g., a pharmaceutically acceptable C 1-5 alkyl or tetrahydrofurfuryl di- or partial-ether of a low molecular weight mono- or poly-oxy-alkanediol.
  • Suitable hydrophilic phase components include, e.g., di- or partial-, especially partial-, -ethers of mono- or poly-, especially mono- or di-, -oxy-alkanediols comprising from 2 to 12, especially 4 carbon atoms.
  • the mono- or poly-oxy-alkanediol moiety is straight-chained.
  • Exemplary hydrophilic phase components for use in relation to the present invention are those known and commercially available under the trade names TRANSCUTOL and COLYCOFUROL. (See U.S. Patent No. 5,342,625).
  • the hydrophilic phase component comprises 1,2- propyleneglycol.
  • the hydrophilic phase component of the present invention may of course additionally include one or more additional ingredients.
  • any additional ingredients will comprise materials in which the vitamin D compound or a mimic thereof is sufficiently soluble, such that the efficacy of the hydrophilic phase as a carrier medium for the vitamin D compound or a mimic thereof is not materially impaired.
  • Examples of possible additional hydrophilic phase components include lower (e.g., C 1-5 ) alkanols, in particular ethanol.
  • compositions of the present invention also comprise one or more surfactants.
  • surfactants that can be used in conjunction with the present invention include hydrophilic or lipophilic surfactants, or mixtures thereof. Especially preferred are non-ionic hydrophilic and non-ionic lipophilic surfactants.
  • Suitable hydrophilic surfactants include reaction products of natural or hydrogenated vegetable oils and ethylene glycol, i.e. polyoxyethylene glycolated natural or hydrogenated vegetable oils, for example polyoxyethylene glycolated natural or hydrogenated castor oils.
  • Such products may be obtained in known manner, e.g., by reaction of a natural or hydrogenated castor oil or fractions thereof with ethylene oxide, e.g., in a molar ratio of from about 1:35 to about 1 :60, with optional removal of free polyethyleneglycol components from the product, e.g., in accordance with the methods disclosed in German Auslegeschriften 1,182,388 and 1,518,819.
  • Suitable hydrophilic surfactants for use in the present pharmaceutical compounds also include polyoxyethylene-sorbitan-fatty acid esters, e.g., mono- and trilauryl, palmityl, stearyl and oleyl esters, e.g., of the type known and commercially available under the trade name TWEEN.
  • polyoxyethylene-sorbitan-fatty acid esters e.g., mono- and trilauryl, palmityl, stearyl and oleyl esters, e.g., of the type known and commercially available under the trade name TWEEN.
  • hydrophilic surfactants for use in the present pharmaceutical compounds are polyoxyethylene alkylethers; polyoxyethylene glycol fatty acid esters, for example polyoxyethylene stearic acid esters; polyglycerol fatty acid esters; polyoxyethylene glycerides; polyoxyethylene vegetable oils; polyoxyethylene hydrogenated vegetable oils; reaction mixtures of polyols and, e.g., fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, and sterols; polyoxyethylene-polyoxypropylene co-polymers; polyoxyethylene-polyoxypropylene block co-polymers; dioctylsuccinate, dioctylsodiumsulfosuccinate, di-[2-ethylhexyl]-succinate or sodium lauryl sulfate; phospholipids, in particular lecithins such as, e.g., soya bean lecithins; propylene glycol mono- and
  • Suitable lipophilic surfactants include alcohols; polyoxyethylene alkylethers; fatty acids; bile acids; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; polyethylene glycol fatty acids esters; polyethylene glycol glycerol fatty acid esters; polypropylene glycol fatty acid esters; polyoxyethylene glycerides; lactic acid esters of mono/diglycerides; propylene glycol diglycerides; sorbitan fatty acid esters; polyoxyethylene sorbitan fatty acid esters; polyoxyethylene-polyoxypropylene block copolymers; trans-esterified vegetable oils; sterols; sugar esters; sugar ethers; sucroglycerides; polyoxyethylene vegetable oils; polyoxyethylene hydrogenated vegetable oils; reaction mixtures of polyols and at least one member of the group consisting of fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils; reaction
  • Suitable lipophilic surfactants for use in the present pharmaceutical compounds also include trans-esterification products of natural vegetable oil triglycerides and polyalkylene polyols.
  • trans-esterification products are known in the art and may be obtained e.g., in accordance with the general procedures described in U.S. Patent No. 3,288,824. They include trans- esterification products of various natural (e.g., non-hydrogenated) vegetable oils for example, maize oil, kernel oil, almond oil, ground nut oil, olive oil and palm oil and mixtures thereof with polyethylene glycols, in particular polyethylene glycols having an average molecular weight of from 200 to 800.
  • polyethylene glycol e.g., having an average molecular weight of from 200 to 800.
  • Various forms of trans-esterification products of the defined class are known and commercially available under the trade name LABRAFIL.
  • Additional surfactants that are suitable for use with the present pharmaceutical compositions include oil-soluble vitamin derivatives, e.g., tocopherol PEG-1000 succinate ("vitamin E TPGS").
  • lipophilic surfactants for use in the present pharmaceutical compounds are mono-, di- and mono/di-glycerides, especially esterification products of caprylic or capric acid with glycerol; sorbitan fatty acid esters; pentaerythritol fatty acid esters and polyalkylene glycol ethers, for example pentaerythrite- -dioleate, -distearate, -monolaurate, -polyglycol ether and -monostearate as well as pentaerythrite-fatty acid esters; monoglycerides, e.g., glycerol monooleate, glycerol monopalmitate and glycerol monostearate; glycerol triacetate or (l,2,3)-triacetin; and sterols and derivatives thereof, for example cholesterols and derivatives thereof, in particular phytosterols, e.g., products comprising sitosterols, e.
  • surfactant compositions contain small to moderate amounts of triglycerides, typically as a result of incomplete reaction of a triglyceride starting material in, for example, a trans-esterification reaction.
  • the surfactants that are suitable for use in the present pharmaceutical compositions include those surfactants that contain a triglyceride.
  • Examples of commercial surfactant compositions containing triglycerides include some members of the surfactant families GELUCIRES, MAISINES, and IMWITORS.
  • GELUCIRE 44/14 saturated polyglycolized glycerides
  • GELUCIRE 50/13 saturated polyglycolized glycerides
  • GELUCIRE 53/10 saturated polyglycolized glycerides
  • GELUCIRE 33/01 saturated polyglycolized glycerides
  • GELUCIRE 39/01 saturated fatty acids
  • GELUCIRE 39/01 synthetic glycerides
  • other GELUCIRES such as 37/06, 43/01, 35/10, 37/02, 46/07, 48/09, 50/02, 62/05, etc.
  • MAISINE 35-1 lainoleic glycerides
  • IMWITOR 742 caprylic/capric glycerides
  • compositions having significant triglyceride content are known to those skilled in the art. It should be appreciated that such compositions, which contain triglycerides as well as surfactants, may be suitable to provide all or part of the lipophilic phase component of the of the present invention, as well as all or part of the surfactants.
  • compositions of the invention will, of course, vary considerably depending on the particular type of composition concerned.
  • the relative proportions will also vary depending on the particular function of ingredients in the composition.
  • the relative proportions will also vary depending on the particular ingredients employed and the desired physical characteristics of the product composition, e.g., in the case of a composition for topical use, whether this is to be a free flowing liquid or a paste. Determination of workable proportions in any particular instance will generally be within the capability of a person of ordinary skill in the art. All indicated proportions and relative weight ranges described below are accordingly to be understood as being indicative of preferred or individually inventive teachings only and not as limiting the invention in its broadest aspect.
  • the lipophilic phase component of the invention will suitably be present in an amount of from about 30% to about 90% by weight based upon the total weight of the composition.
  • the lipophilic phase component is present in an amount of from about 50% to about 85% by weight based upon the total weight of the composition.
  • the surfactant or surfactants of the invention will suitably be present in an amount of from about 1% to 50% by weight based upon the total weight of the composition.
  • the surfactant(s) is present in an amount of from about 5% to about 40% by weight based upon the total weight of the composition.
  • the amount of vitamin D compound or a mimic thereof in compositions of the invention will of course vary, e.g., depending on the intended route of administration and to what extent other components are present. In general, however, the vitamin D compound or a mimic thereof of the invention will suitably be present in an amount of from about 0.005% to 20% by weight based upon the total weight of the composition. Preferably, the vitamin D compound or a mimic thereof, is present in an amount of from about 0.01% to 15% by weight based upon the total weight of the composition.
  • the hydrophilic phase component of the invention will suitably be present in an amount of from about 2% to about 20% by weight based upon the total weight of the composition.
  • the hydrophilic phase component is present in an amount of from about 5% to 15% by weight based upon the total weight of the composition.
  • the pharmaceutical composition of the invention may be in a semisolid formulation.
  • Semisolid formulations within the scope of the invention may comprise, e.g., a lipophilic phase component present in an amount of from about 60% to about 80% by weight based upon the total weight of the composition, a surfactant present in an amount of from about 5% to about 35% by weight based upon the total weight of the composition, and a vitamin D compound or a mimic thereof, present in an amount of from about 0.01% to about 15% by weight based upon the total weight of the composition.
  • compositions of the invention may be in a liquid formulation.
  • Liquid formulations within the scope of the invention may comprise, e.g., a lipophilic phase component present in an amount of from about 50% to about 60% by weight based upon the total weight of the composition, a surfactant present in an amount of from about 4% to about 25% by weight based upon the total weight of the composition, a vitamin D compound or a mimic thereof present in an amount of from about 0.01% to about 15% by weight based upon the total weight of the composition, and a hydrophilic phase component present in an amount of from about 5% to about 10% by weight based upon the total weight of the composition.
  • compositions that may be used include the following, wherein the percentage of each component is by weight based upon the total weight of the composition excluding the vitamin D compound or a mimic thereof: Gelucire 44/14 about 50% Miglyol 812 about 50%;
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 5% Miglyol 812 about 65%;
  • Vitamin E TPGS about 20%
  • Vitamin E TPGS about 30%
  • Vitamin E TPGS about 25% Miglyol 812 about 15%;
  • Vitamin E TPGS about 50% PEG 4000 about 50%;
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 5% Miglyol 812 about 95%; aa. Vitamin E TPGS about 5% Miglyol 812 about 65% PEG 4000 about 30%;
  • Vitamin E TPGS about 10% Miglyol 812 about 90%;
  • Vitamin E TPGS about 5% Miglyol 812 about 85% PEG 4000 about 10%;
  • Vitamin E TPGS about 10%
  • Miglyol 812 about 80%
  • the pharmaceutical compositions comprise a vitamin D compound or a mimic thereof, a lipophilic component, and a surfactant.
  • the lipophilic component may be present in any percentage from about 1% to about 100%.
  • the lipophilic component may be present at about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93
  • the surfactant may be present in any percentage from about 1% to about 100%.
  • the surfactant may be present at about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%.
  • the lipophilic component is MIGLYOL 812 and the surfactant is vitamin E TPGS.
  • the pharmaceutical compositions comprise about 50% MIGLYOL 812 and about 50% vitamin E TPGS, about 90% MIGLYOL 812 and about 10% vitamin E TPGS, or about 95% MIGLYOL 812 and about 5% vitamin E TPGS.
  • the pharmaceutical compositions comprise a vitamin D compound and a lipophilic component, e.g., around 100% MIGLYOL 812.
  • the pharmaceutical compositions comprise about 50% MIGLYOL 812, about 50% vitamin E TPGS, and small amounts of BHA and BHT.
  • This formulation has been shown to be unexpectedly stable, both chemically and physically (see published U.S. Appln. 2003/0191093).
  • the enhanced stability provides the compositions with a longer shelf life.
  • the stability also allows the compositions to be stored at room temperature, thereby avoiding the complication and cost of storage under refrigeration.
  • this composition is suitable for oral administration and has been shown to be capable of solubilizing high doses of vitamin D compound, thereby enabling high dose pulse administration of vitamin D compounds for the treatment of hyperproliferative diseases and other disorders.
  • the pharmaceutical compositions comprise about 50% MIGLYOL 812, about 50% vitamin E TPGS, and about 0.01% to about 0.50% each of BHA and BHT. In other embodiments, the pharmaceutical compositions comprise about 50% MIGLYOL 812, about 50% vitamin E TPGS, and about 0.05% to about 0.35% each of BHA and BHT. In certain embodiments, the pharmaceutical compositions comprise about 50% MIGLYOL 812, about 50% vitamin E TPGS, about 0.35% BHA, and about 0.10% BHT.
  • compositions that may be used include the following, wherein the percentage of each component is by weight based upon the total weight of the composition excluding the vitamin D compound or a mimic thereof:
  • BHA about 0.05% BHT about 0.05%
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 50%
  • Vitamin E TPGS about 50%
  • BHT about 0.08%. It will be understood by those of skill in the art that the formulations of the invention comprising a lipophilic component and a surfactant in amounts that total about 100% (e.g., about 50% lipophilic component and about 50% surfactant) provide adequate room for the vitamin D compound and additives (e.g., antioxidants) which are present in the formulation in small amounts, each generally present at less than 1% by weight.
  • a lipophilic component and a surfactant e.g., about 50% lipophilic component and about 50% surfactant
  • compositions comprising the vitamin D compound of the present invention may further comprise one or more additives.
  • Additives that are well known in the art include, e.g., detackifiers, anti- foaming agents, buffering agents, antioxidants (e.g., ascorbyl palmitate, butyl hydroxy anisole (BHA), butyl hydroxy toluene (BHT) and tocopherols, e.g., ⁇ - tocopherol (vitamin E)), preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
  • antioxidants e.g., ascorbyl palmitate, butyl hydroxy anisole (BHA), butyl hydroxy toluene (BHT) and tocopherols, e.g., ⁇ - tocopherol (vitamin E)
  • antioxidants such as BHA and BHT may each be present in an amount of from about 0.01% to about 0.50% by weight based upon the total weight of the composition, e.g., about 0.05 to about 0.35% by weight, e.g., about 0.01, 0.05, 0.10, 0.15, 0.20, 0.25, 0.30, 0.35, 0.40, 0.45, or 0.50% by weight.
  • the additive may also comprise a thickening agent.
  • suitable thickening agents may be those known and employed in the art, including, e.g., pharmaceutically acceptable polymeric materials and inorganic thickening agents.
  • Exemplary thickening agents for use in the present pharmaceutical compositions include polyacrylate and polyacrylate copolymer resins, for example poly-acrylic acid and poly-acrylic acid/methacrylic acid resins; celluloses and cellulose derivatives including: alkyl celluloses, e.g., methyl-, ethyl- and propyl-celluloses; hydroxyalkyl- celluloses, e.g., hydroxypropyl-celluloses and hydroxypropylalkyl-celluloses such as hydroxypropyl-methyl-celluloses; acylated celluloses, e.g., cellulose- acetates, cellulose-acetatephthallates, cellulose-acetatesuccinates and hydroxypropylmethyl-cellulose phthallates; and salts thereof such
  • Such thickening agents as described above may be included, e.g., to provide a sustained release effect.
  • the use of thickening agents as aforesaid will generally not be required and is generally less preferred.
  • Use of thickening agents is, on the other hand, indicated, e.g., where topical application is foreseen.
  • compositions in accordance with the present invention may be employed for administration in any appropriate manner, e.g., orally, e.g., in unit dosage form, for example in a solution, in hard or soft encapsulated form including gelatin encapsulated form, parenterally or topically, e.g., for application to the skin, for example in the form of a cream, paste, lotion, gel, ointment, poultice, cataplasm, plaster, dermal patch or the like, as a coating for a medical device, e.g., a stent, or for ophthalmic application, for example in the form of an eye-drop, -lotion or -gel formulation.
  • Readily flowable forms, for example solutions and emulsions may also be employed e.g., for intralesional injection, or may be administered rectally, e.g., as an enema.
  • the vitamin D compound When the composition of the present invention is formulated in unit dosage form, the vitamin D compound will preferably be present in an amount of between 1 and 1000 ⁇ g per unit dose. More preferably, the amount of vitamin D compound per unit dose will be about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 ⁇ g or any amount therein.
  • the amount of vitamin D compound per unit dose will be about 5 ⁇ g to about 180 ⁇ g, more preferably about 10 ⁇ g to about 135 ⁇ g, more preferably about 45 ⁇ g.
  • the unit dosage form comprises 45, 90, 135, or 180 ⁇ g of calcitriol.
  • the total quantity of ingredients present in the capsule is preferably about 10-1000 ⁇ L. More preferably, the total quantity of ingredients present in the capsule is about 100-300 ⁇ L. In another embodiment, the total quantity of ingredients present in the capsule is preferably about 10-1500 mg, preferably about 100- 1000 mg. In one embodiment, the total quantity is about 225, 450, 675, or 900 mg.
  • the unit dosage form is a capsule comprising 45, 90, 135, or 180 ⁇ g of calcitriol.
  • Animals which may be treated according to the present invention include all animals which may benefit from administration of the compounds of the present invention. Such animals include humans, pets such as dogs and cats, and veterinary animals such as cows, pigs, sheep, goats and the like.
  • Tumor blood vessels are assessed by CD31 immunohistochemistry and hypoxia regions are identified by uptake of EF5.
  • Composite images of doxorubicin, CD31, and EF5 fluorescence are generated using Media Cybernetics Image Pro PLUS and scanned for pixel density. Characteristic penetration lengths are determined and plotted as a function of distance to the nearest CD31 + blood vessel.
  • mice are implanted subcutaneously with 1 x 10 6 PC-3 prostate tumor cells in 0.2 ml Matrigel ® . After the tumor volume reaches 1000 mm 3 , mice are treated with 35 ⁇ g/kg calcitriol on days 1, 2, and 3. The effects of high dose calcitriol treatment on tumor blood flow are monitored using high-frequency micro-ultrasound functional imaging at 24-72 hours following calcitriol treatment. Tumors are monitored for markers of perfusion using Hoescht staining and for hypoxia by piminozadole staining.
  • CRP HR 1.38, 95% CI 1.15-1.66, p ⁇ 0.001

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Obesity (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
PCT/US2007/023030 2006-11-01 2007-11-01 Use of vitamin d derivatives to enhance delivery of therapeutics and oxygen to tumors WO2008057363A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US86390906P 2006-11-01 2006-11-01
US60/863,909 2006-11-01
US96816007P 2007-08-27 2007-08-27
US60/968,160 2007-08-27

Publications (2)

Publication Number Publication Date
WO2008057363A2 true WO2008057363A2 (en) 2008-05-15
WO2008057363A3 WO2008057363A3 (en) 2008-08-14

Family

ID=39365038

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/023030 WO2008057363A2 (en) 2006-11-01 2007-11-01 Use of vitamin d derivatives to enhance delivery of therapeutics and oxygen to tumors

Country Status (3)

Country Link
CL (1) CL2007003162A1 (es)
TW (1) TW200833345A (es)
WO (1) WO2008057363A2 (es)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9872866B2 (en) 2013-04-24 2018-01-23 Salk Institute For Biological Studies Vitamin D receptor/SMAD genomic circuit gates fibrotic response
US9895381B2 (en) 2013-06-05 2018-02-20 Salk Institute For Biological Studies Vitamin D receptor agonists to treat diseases involving CXCL12 activity
EP2695617B1 (en) * 2009-12-23 2019-01-09 Glycomyr Inc. Vitamin D glycosides and sulfates for treating intestinal diseases
EP2861234B1 (en) 2012-06-18 2019-03-27 TherapeuticsMD, Inc. Progesterone formulations
US11376264B2 (en) 2017-07-24 2022-07-05 Salk Institute For Biological Studies Use of bromodomain-containing protein 9 antagonists in combination with vitamin D receptor agonists in diabetes treatment
US11654300B2 (en) 2020-01-28 2023-05-23 Reflexion Medical, Inc. Joint optimization of radionuclide and external beam radiotherapy

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5654333A (en) * 1991-10-21 1997-08-05 The United States Of America As Represented By The Department Of Health And Human Services Methods for prevention of cancer using phenylacetic acids and derivatives thereof
US6503893B2 (en) * 1996-12-30 2003-01-07 Bone Care International, Inc. Method of treating hyperproliferative diseases using active vitamin D analogues
US6521608B1 (en) * 1998-03-27 2003-02-18 Oregon Health & Science University Vitamin D and its analogs in the treatment of tumors and other hyperproliferative disorders
US20040191263A1 (en) * 2001-07-27 2004-09-30 Hageman Robert Johan Joseph Enteral compositions for the prevention and/or treatment of sepsis
US20040253323A1 (en) * 2003-06-11 2004-12-16 Giles Brian C. Ionic cancer therapy and methods for using same in the treatment of tumors and metastasis
US20060078494A1 (en) * 2004-09-24 2006-04-13 Sapphire Therapeutics, Inc. Use of inhibitors of 24-hydroxylase in the treatment of cancer

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5654333A (en) * 1991-10-21 1997-08-05 The United States Of America As Represented By The Department Of Health And Human Services Methods for prevention of cancer using phenylacetic acids and derivatives thereof
US6503893B2 (en) * 1996-12-30 2003-01-07 Bone Care International, Inc. Method of treating hyperproliferative diseases using active vitamin D analogues
US6521608B1 (en) * 1998-03-27 2003-02-18 Oregon Health & Science University Vitamin D and its analogs in the treatment of tumors and other hyperproliferative disorders
US20040191263A1 (en) * 2001-07-27 2004-09-30 Hageman Robert Johan Joseph Enteral compositions for the prevention and/or treatment of sepsis
US20040253323A1 (en) * 2003-06-11 2004-12-16 Giles Brian C. Ionic cancer therapy and methods for using same in the treatment of tumors and metastasis
US20060078494A1 (en) * 2004-09-24 2006-04-13 Sapphire Therapeutics, Inc. Use of inhibitors of 24-hydroxylase in the treatment of cancer

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2695617B1 (en) * 2009-12-23 2019-01-09 Glycomyr Inc. Vitamin D glycosides and sulfates for treating intestinal diseases
EP2861234B1 (en) 2012-06-18 2019-03-27 TherapeuticsMD, Inc. Progesterone formulations
EP3560500B1 (en) 2012-06-18 2021-12-22 TherapeuticsMD, Inc. Progesterone formulations
US9872866B2 (en) 2013-04-24 2018-01-23 Salk Institute For Biological Studies Vitamin D receptor/SMAD genomic circuit gates fibrotic response
US10238667B2 (en) 2013-04-24 2019-03-26 Salk Institute For Biological Studies Vitamin D receptor/SMAD genomic circuit gates fibrotic response
US9895381B2 (en) 2013-06-05 2018-02-20 Salk Institute For Biological Studies Vitamin D receptor agonists to treat diseases involving CXCL12 activity
US11376264B2 (en) 2017-07-24 2022-07-05 Salk Institute For Biological Studies Use of bromodomain-containing protein 9 antagonists in combination with vitamin D receptor agonists in diabetes treatment
US11654300B2 (en) 2020-01-28 2023-05-23 Reflexion Medical, Inc. Joint optimization of radionuclide and external beam radiotherapy

Also Published As

Publication number Publication date
CL2007003162A1 (es) 2008-05-30
WO2008057363A3 (en) 2008-08-14
TW200833345A (en) 2008-08-16

Similar Documents

Publication Publication Date Title
US20060172014A1 (en) Treatment of lung cancer with active vitamin D compounds in combination with other treatments
US20070275934A1 (en) Treatment of pancreatic cancer with active vitamin d compounds in combination with other treatments
US20060177374A1 (en) Treatment of cancer with active vitamin D compounds in combination with radiotherapeutic agents and treatments
US20070004688A1 (en) Pharmaceutical compositions comprising active vitamin D compounds
Markman et al. Phase I safety, pharmacokinetic, and pharmacodynamic study of the oral phosphatidylinositol-3-kinase and mTOR inhibitor BGT226 in patients with advanced solid tumors
WO2008057363A2 (en) Use of vitamin d derivatives to enhance delivery of therapeutics and oxygen to tumors
CA2528552A1 (en) Pharmaceutical compositions comprising active vitamin d compounds
US20060189586A1 (en) Pharmaceutical compositions comprising active vitamin D compounds
US20070142339A1 (en) Prevention of arterial restenosis with active vitamin d compounds
TWI729394B (zh) 雄性激素受體調節劑及其用途
US8501717B2 (en) Methods to treat and/or prevent mucositis
US20040001835A1 (en) Prevention or treatment of cancer using integrin alphavbeta3 antagonists in combination with other agents
US20090069276A1 (en) Treatment, prevention and amelioration of pulmonary disorders associated with chemotherapy or radiotherapy with active vitamin D compounds or mimics thereof
EP1833485A2 (en) Prevention of thrombotic disorders with active vitamin d compounds or mimics thereof
US20080069814A1 (en) Prevention of Thrombotic Disorders with Active Vitamin D Compounds or Mimics Thereof
CN102448466A (zh) 伊奈骨化醇的新治疗应用
ZA200510025B (en) Pharmaceutical compositions comprising active vitamin D compounds
CN101163483A (zh) 使用活性维生素d化合物或其模拟物治疗、预防和缓解与化疗和放疗有关的肺部疾患
Jang et al. Skeletal-Related Events in Patients with Metastatic Renal Cell Carcinoma: A Systematic Review

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07861618

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07861618

Country of ref document: EP

Kind code of ref document: A2