WO2004065572A2 - Proteic binding partners of tctp and methods of modulating tumor reversion or cell apoptosis - Google Patents

Proteic binding partners of tctp and methods of modulating tumor reversion or cell apoptosis Download PDF

Info

Publication number
WO2004065572A2
WO2004065572A2 PCT/US2004/001894 US2004001894W WO2004065572A2 WO 2004065572 A2 WO2004065572 A2 WO 2004065572A2 US 2004001894 W US2004001894 W US 2004001894W WO 2004065572 A2 WO2004065572 A2 WO 2004065572A2
Authority
WO
WIPO (PCT)
Prior art keywords
tctp
binding partner
cell
tumor
compound
Prior art date
Application number
PCT/US2004/001894
Other languages
French (fr)
Other versions
WO2004065572A3 (en
Inventor
Adam Telerman
Robert Amson
Brent Passer
Original Assignee
Molecular Engines Laboratories
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 Molecular Engines Laboratories filed Critical Molecular Engines Laboratories
Publication of WO2004065572A2 publication Critical patent/WO2004065572A2/en
Publication of WO2004065572A3 publication Critical patent/WO2004065572A3/en
Priority to US11/187,891 priority Critical patent/US20060140970A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention encompasses methods of modulating tumor reversion or cell apoptosis. Also encompassed are methods of identifying compounds useful for treating, preventing and managing cancer or a neurodegenerative disorder.
  • Cancer is characterized primarily by an increase in the number of abnormal cells derived from a given normal tissue, invasion of adjacent tissues by these abnormal cells, or lymphatic or blood-borne spread of malignant cells to regional lymph nodes and to distant sites (metastasis).
  • Clinical data and molecular biologic studies indicate that cancer is a multi-step that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia.
  • the neoplastic region may evolve clonically and develop an increasing capacity for invasion, growth, metastasis, and heterogenicity, especially under conditions in which neoplastic cells escape the host's immune surveillance.
  • Tumor reversion is a process wherein malignant cells revert to more normal phenotypes.
  • the study of tumor reversion has identified genes that are differentially expressed between tumor cells and their revertants. Tuynder et al., Proc. Natl. Acad. Sci. USA, 99(23): 14976-14981 (2002).
  • One of those is the gene tptl, which produces the Translationally Controlled Tumor Protein ("TCTP”), which has been found to be overexpressed in tumor cells, but not in the revertants.
  • TCTP Translationally Controlled Tumor Protein
  • neurodegenerative disorders are characterized by progressive and irreversible loss of neurons from specific regions of the brain. As a group, these disorders are relatively common and represent a substantial medical and social problem.
  • This invention is based, in part, on the inventors' discovery that certain proteins, i.e., proteic binding partners of TCTP, interact with TCTP, and modulate (i.e., enhance/promote or reduce/inhibit) tumor reversion and/or cell apoptosis. Accordingly, this invention encompasses a method of modulating tumor reversion and/or cell apoptosis comprising contacting a tumor cell with an effective amount of a proteic binding partner of TCTP.
  • This invention also encompasses a method of identifying a compound that reduces or inhibits the binding between TCTP and a proteic binding partner of TCTP. Compounds so identified can be further screened for their efficacy in promoting or reducing/inhibiting tumor reversion or cell apoptosis.
  • proteic binding partners of TCTP can be directly used for the treatment, prevention or management. Some proteic binding partners of TCTP, which promotes tumor reversion or cell apoptosis, can be used for the treatment, prevention or management of cancer, while other proteic binding partners of TCTP, which reduces or inhibits tumor reversion or cell apoptosis, can be used for the treatment, prevention or management of a neurodegenerative disorder. In other embodiments, other compounds, selected using methods based on the interaction between TCTP and its proteic binding partners, are used for the treatment, prevention or management of cancer or a neurodegenerative disorder. 4. BRIEF DESCRIPTION OF FIGURES
  • Figure 1 illustrates the inhibition of GTP-GDP exchange activity of Arfl- GAP by TCTP.
  • TCTP is a ubiquitously expressed protein that is evolutionarily conserved. Its role in tumor reversion and/or cell apoptosis has been implicated. Tuynder et al., Proc. Natl. Acad. Sci. USA, 99(23): 14976-81 (2002).
  • This invention is based, in part, on the inventors' discovery that certain proteins, i.e., proteic binding partners of TCTP, can interact with TCTP, and thus may be used to modulate TCTP's effect on tumor reversion and/or cell apoptosis.
  • a first embodiment of this invention encompasses methods of modulating tumor reversion and/or cell apoptosis comprising contacting a tumor cell with an effective amount of a proteic binding partner of TCTP.
  • modulating when used in connection with tumor reversion or cell apoptosis, mean that the extent or intensity of such tumor reversion or cell apoptosis is altered or adjusted.
  • the terms encompass increase, promotion, decrease, reduction and inhibition of tumor reversion or cell apoptosis.
  • promote promote
  • tumor reversion or cell apoptosis when used in connection with tumor reversion or cell apoptosis, mean that tumor reversion or cell apoptosis in a cell, treated using methods of this invention, is higher than the same type of cell without the treatment. In some embodiments, tumor reversion or cell apoptosis in a cell treated using methods of this invention is higher than that in the same type of cell without the treatment by about 10 percent, 30 percent, 50 percent, 70 percent, 100 percent, 200 percent or more.
  • the terms “decrease,” “reduce,” and “reduction,” when used in connection with tumor reversion or cell apoptosis means that tumor reversion or cell apoptosis in a cell, treated using methods of this invention, is lower than the same type of cell without the treatment. In some embodiments, tumor reversion or cell apoptosis in a cell treated using methods of this invention is lower than that in the same type of cell without the treatment by about 10 percent, 30 percent, 50 percent, 70 percent or 90 percent or more. In this regard, the terms “inhibit” and “inhibition” mean that tumor reversion or cell apoptosis in a cell treated using methods of this invention is substantially abolished.
  • tumor reversion or cell apoptosis is inhibited where no or little such tumor reversion or cell apoptosis can be detected using a well-known analytical method such as SDS-PAGE, PARP cleavage, spectrophotometry, autoradiography, or chromatographic techniques.
  • proteic binding partner of TCTP means a protein, or portions thereof, that can bind to or interact with TCTP. Any known methods of identification of a binding partner may be used, e.g., co- precipitation and fluorescence resonance energy transfer. A specific method is yeast two- hybrid system, derived from the system described by Finley and Brent (Interaction trap cloning with yeast, in DNA Cloning, Expression Systems: A Practical Approach, pp. 169-203, Oxford Universal Press, Oxford (1995), which is incorporated herein by reference). Examples of proteic binding partners of TCTP include, but are not limited to, human elongation factor- 1 Delta and those listed in Table 1.
  • this invention encompasses methods of identifying compounds that reduce or inhibit binding between TCTP and a proteic binding partner of TCTP comprising: (a) contacting a compound with TCTP and the proteic binding partner of TCTP; and (b) determining whether the binding between TCTP and the proteic binding partner of TCTP is decreased.
  • the terms “reduce” and “decrease,” when used in connection with binding, mean that the binding between two molecules are impeded, slowed or prevented in the presence of a test compound as compared to the binding occurring in the absence of such a test compound. Binding between two molecules may be determined by any methods known in the art, including, but not limited to, fluorescence, spectroscopy, chromatography, centrifugation and electrophoresis.
  • binding between TCTP and a proteic binding partner of TCTP is reduced by about 10 percent, 20 percent, 40 percent, 60 percent, 80 percent or 90 percent or more in the presence of the test compound as compared to the binding of the same molecules in the absence of the test compound.
  • this invention encompasses methods of identifying compounds that are useful in treating, preventing or managing cancer.
  • One aspect of this embodiment encompasses methods of identifying compounds that promote tumor reversion or cell apoptosis comprising: (a) contacting a proteic binding partner of TCTP with a tumor cell; and (b) determining whether tumor reversion or cell apoptosis is increased as compared to a tumor cell that has not been contacted with the proteic binding partner of TCTP.
  • the proteic binding partner identified positive using the methods can be used in treating, preventing or managing cancer.
  • this invention also encompasses methods of identifying compounds that promote tumor reversion or cell apoptosis comprising: (a) contacting a compound with TCTP and a proteic binding partner of TCTP; (b) determining whether binding between TCTP and the proteic binding partner of TCTP is decreased; and wherein the binding between TCTP and the proteic binding partner of TCTP is decreased, then (c) contacting the compound with a tumor cell; and (d) determining whether tumor reversion or cell apoptosis is increased as compared to a tumor cell that has not been contacted with the compound.
  • Compounds tested positive using this method is potentially useful for the treatment, prevention and/or management of cancer.
  • this invention also encompasses methods of identifying compounds potentially useful in the treatment, prevention and/or management of a neurodegenerative disorder such as Alzheimer's disease.
  • One aspect of this embodiment encompasses methods of identifying compounds that reduce or inhibit tumor reversion or cell apoptosis comprising: (a) contacting a proteic binding partner of TCTP with a tumor cell; and (b) determining whether tumor reversion or cell apoptosis is reduced as compared to a tumor cell that has not been contacted with the proteic binding partner of TCTP.
  • this invention also encompasses methods of identifying compounds that reduce or inhibit tumor reversion or cell apoptosis comprising: (a) contacting a compound with TCTP and a proteic binding partner of TCTP; (b) determining whether binding between TCTP and the proteic binding partner of TCTP is decreased; and wherein the binding between TCTP and the proteic binding partner of TCTP is decreased, then (c) contacting the compound with a tumor cell; and (d) determining whether tumor reversion or cell apoptosis is reduced as compared to a tumor cell that has not been contacted with the compound.
  • tumor reversion or cell apoptosis increases or decreases in the tumor cell contacted with the test compound can be determined by any methods known in the art, as well as those described herein. The determination can be done in vivo or in vitro. For example, where the tests are carried out in vitro, models such as K256/KS cells described in Telerman et al., Proc. Natl. Acad. Sci. USA, 90: 8702-6 (1993), Ml-LTR cells described in Amson et al., Proc. Natl. Acad. Sci. USA, 93: 3953-7 (1996), and U937/US3-US4 cells described in Nemani et al., Proc. Natl. Acad. Sci.
  • test compound in the case of in vivo tests, can be injected into an animal model, such as immunodepressed mice, and effects of the injected test compound may be investigated. Specific conditions and thresholds for identification are well within the ordinary skill in the art.
  • PARP Poly ADP-Ribose Polymerase
  • this invention encompasses methods of treating, preventing and/or managing cancer comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of a compound identified using certain methods of this invention.
  • the terms “treat,” “treating “ and “treatment” refer to the eradication or amelioration of a disease or condition, or of one or more symptoms associated with the disease or condition. In certain embodiments, the terms refer to minimizing the spread or worsening of the disease or condition resulting from the administration of one or more prophylactic or therapeutic agents to a subject with such a disease or condition. As used herein, and unless otherwise specified, the terms “prevent,”
  • preventing and prevention refer to the prevention of the onset, recurrence or spread of a disease or condition, or of a symptom thereof.
  • the terms “manage,” “managing” and “management” refer to preventing or slowing the progression, spread or worsening of a disease or condition. Often, the beneficial effects that a subject derives from a prophylactic or therapeutic agent do not result in a cure of the disease or condition.
  • the term "managing” encompasses preventing the recurrence of cancer in a patient who had suffered from cancer, lengthening the time a patient who had suffered from cancer remains in remission, preventing the occurrence of cancer in patients at risk of suffering from cancer (e.g., patients who had been exposed to high amounts of radiation or carcinogenic materials, such as asbestos; patients infected with viruses associated with the occurrence of cancer, such as, but not limited to, HIV and Kaposi's sarcoma-associated herpes virus; and patients with genetic predispositions to cancer, such as those suffering from Downs syndrome), and preventing the occurrence of malignant cancer in patients suffering from pre-malignant or non-malignant cancers.
  • cancer e.g., patients who had been exposed to high amounts of radiation or carcinogenic materials, such as asbestos; patients infected with viruses associated with the occurrence of cancer, such as, but not limited to, HIV and Kaposi's sarcoma-associated herpes virus; and patients with genetic predispositions
  • a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment or management of a disease or condition, or to delay or minimize one or more symptoms associated with the disease or condition.
  • a therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other therapies, which provides a therapeutic benefit in the treatment or management of the disease or condition.
  • the term "therapeutically effective amount” can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease or condition, or enhances the therapeutic efficacy of another therapeutic agent.
  • a prophylactically effective amount of a compound is an amount sufficient to prevent a disease or condition, or prevent its recurrence.
  • a prophylactically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other agents, which provides a prophylactic benefit in the prevention of the disease.
  • the term “prophylactically effective amount” can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent.
  • cancer includes, but is not limited to, solid tumors and blood born tumors.
  • cancer refers to disease of skin tissues, organs, blood, and vessels, including, but not limited to, cancers of the bladder, bone or blood, brain, breast, cervix, chest, colon, endrometrium, esophagus, eye, head, kidney, liver, lymph nodes, lung, mouth, neck, ovaries, nancreas, prostate, rectum, stomach, testis, throat, and uterus.
  • Specific cancers include, but are not limited to, advanced malignancy, amyloidosis, neuroblastoma, meningioma, hemangiopericytoma, multiple brain metastases, glioblastoma multiforms, glioblastoma, brain stem glioma, poor prognosis malignant brain tumor, malignant glioma, recurrent malignant giolma, anaplastic astrocytoma, anaplastic oligodendroglioma, neuroendocrine tumor, rectal adenocarcinoma, Dukes C & D colorectal cancer, unresectable colorectal carcinoma, metastatic hepatocellular carcinoma, Kaposi's sarcoma, karotype acute myeloblastic leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell
  • compounds of this invention can be administered, sequentially or simultaneously, with another anticancer agent.
  • the administration may be via same route or different routes.
  • anti-cancer agents include, but are not limited to: acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carbop
  • anti-cancer drugs include, but are not limited to: 20-epi-l,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morpho genetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PT
  • cartilage derived inliibitor carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin
  • a prophylactic or therapeutic dose of each active ingredient in the treatment, prevention and/or management of cancer will typically vary with the specific active ingredients, the severity and type of cancer, and the route of administration.
  • the dose, and perhaps the dose frequency may also vary according to age, body weight, response, and the past medical history of the patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors by following, for example, dosages reported in the literature and recommended in the Physician 's Desk Reference ® (54 th ed., 2000).
  • the patient is an animal. In another embodiment, the patient is a mammal, specifically, a human.
  • this invention encompasses methods of treating, preventing and managing a neurodegenerative disorder comprising administering to a patient in need of such treatment, prevention and/or management a therapeutically or prophylactically effective amount of a compound identified certain methods of this invention.
  • neurodegenerative disorders examples include, but are not limited to, amylotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease and Parkinson's disease.
  • the neurodegenerative disorder that can be treated, prevented and/or managed using methods of this invention is Alzheimer's disease, hi one embodiment, the patient is a mammal, in particular, a human.
  • compound of this invention may be administered, sequentially or simultaneously, with another agent useful in treating a neurodegenerative disorder. The administration may be via same route or different routes.
  • agents include, but are not limited to: acridine derivatives such as tacrine; amantadine; dopamine depleting agents such as tetrabenazine and reserpine; dopamine receptor antagonists such as bromocriptine and pergolide; metabolic precursors of dopamine such as levodopa; inhibitors of acetylcholinesterase (AChE) such as physostigmine; inhibitors of L-amino acid decarboxylase such as benserazide and carbidopa; muscarinic receptor antagonists such as benzotropine mesylate, diphenhydramine hydorchloride and trihexyphenidyl; precursors of acetylcholine synthesis such as choline chloride and phosphatidyl choline (lecithin); and selegiline.
  • acridine derivatives such as tacrine
  • amantadine dopamine depleting agents such as
  • a prophylactic or therapeutic dose of each active ingredient in the treatment, prevention and/or management of a neurodegenerative disorder will typically vary with the specific active ingredients, the severity and type of disorder, and the route of administration.
  • the dose, and perhaps the dose frequency may also vary according to age, body weight, response, and the past medical history of the patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors by following, for example, dosages reported in the literature and recommended in the Physician 's Desk Reference ® (54 m ed., 2000).
  • this invention encompasses pharmaceutical compositions comprising one or more proteic binding partners of TCTP, or genes thereof.
  • this invention also encompasses pharmaceutical compositions comprising one or more of compounds identified using methods of this invention. Such pharmaceutical compositions are described below.
  • compositions are single unit dosage forms suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial), or transdermal administration to a patient.
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial
  • transdermal administration e.g., transdermal administration to a patient.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols ( e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g., aque
  • the formulation should suit the mode of administration.
  • oral administration requires enteric coatings to protect the compounds of this invention from degradation within the gastrointestinal tract.
  • the compounds of this invention may be administered in a liposomal formulation to shield the compounds from degradative enzymes, facilitate transport in circulatory system, and effect delivery across cell membranes to intracellular sites.
  • composition, shape, and type of dosage forms of the invention will typically vary depending on their use.
  • a dosage form used in the acute treatment of a disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease.
  • compositions of the invention that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
  • Typical oral dosage forms of the invention are prepared by combining the active ingredients in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
  • Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • Disintegrants or lubricants can be used in pharmaceutical compositions and dosage forms of the invention.
  • Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art.
  • Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, 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 such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl rnyristate, and benzyl benzoate.
  • aqueous vehicles such as, 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 such as, but not limited to, ethyl alcohol, poly
  • Transdermal, topical, and mucosal dosage forms of the invention include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington 's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980 & 1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985).
  • Transdermal dosage forms include "reservoir type” or "matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • additional components may be used prior to, in conjunction with, or subsequent to treatment with active ingredients of the invention.
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • lactose-free means that the amount of lactose present, if any, is insufficient to substantially increase the degradation rate of an active ingredient.
  • Lactose-free compositions of the invention can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) 25-NF20 (2002).
  • lactose-free compositions comprise active ingredients, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • Preferred lactose-free dosage forms comprise active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80.
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anliydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained.
  • anhydrous compositions are preferably packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients.
  • Active ingredients of the invention can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is incorporated herein by reference.
  • Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microp articles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the compounds of this invention.
  • the invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • Active ingredients in pharmaceutical compositions of this invention may be in the form of nucleic acids. Delivery of these molecules into a patient can either be direct, i.e., the patient is directly exposed to the compounds of this invention or compound-carrying vector, or indirect, i.e., cells are first transformed with the compounds of this invention in vitro, then transplanted into the patient for cell replacement therapy. These two approaches are known as in vivo and ex vivo therapy, respectively. In the case of in vivo therapy, compounds of this invention are directly administered in vivo, where they are expressed to produce the encoded product.
  • compounds of this invention can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor, as disclosed in, for example, WO 92/06180, WO 92/22635, WO 92/20316, WO 93/14188 and WO 93/20221, all of which are incorporated herein by reference.
  • Ex vivo therapy involves transferring the compounds of this invention to cells in tissue culture by methods such as electroporation, lipofection, calcium phosphate mediated transfection and viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells.
  • the r.ells are then placed under selection to isolate those cells that have taken up and are expressing the transferred compounds. These cells are then delivered to a patient.
  • Compounds of this invention are introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any methods known in the art, including, but not limited to, transfection, electroporation, microinjection, infection with a viral vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer and spheroplast fusion.
  • Numerous techniques are known in the art for the introduction of foreign compounds into cells. Examples of such techniques are disclosed in: Loeffler et al, Meth. Enzymol, 217: 599-618 (1993); Cohen et al, Meth. Enzymol, 217: 618-644 (1993); and Cline, Pharmc.
  • the resulting recombinant cells can be delivered to a patient by various methods known in the art. Examples of the delivery methods include, but are not limited to, subcutaneous injection, skin graft and intravenous injection.
  • kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a patient.
  • a typical kit of the invention comprises a single unit dosage form of the compounds of this invention, or a pharmaceutically acceptable salt, prodrug, solvate, hydrate, clathrate or stereoisomer thereof, and a single unit dosage form of another agent that may be used in combination with the compounds of this invention.
  • Eats of the invention can further comprise devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, drip bags, patches, and inhalers.
  • Kits of the invention can further comprise pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients.
  • the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration.
  • Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, 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 such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, 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 such as, but not limited to, ethyl alcohol
  • the results show that the function of Arfl , a binding partner of TCTP, is affected by the presence of TCTP.
  • TCTP inhibits the GTP-GDP exchange catalyzed by Arfl -GAP, implicating that Arfl is kept in its active conformation in presence of TCTP.
  • Radio-labeled TCTP is generated from an in vitro translation in a rabbit reticulocyte lysate in the presence of 35 S-labeled methionine and 35 S-labeled cysteine.
  • GST-fusion protein of a binding partner of TCTP is prepared, and is coupled to sepharose-glutathione beads.
  • 35 S labeled TCTP, sepharose-glutathione coupled binding partner and a test compound are mixed and incubated for a sufficient time.
  • a mixture without the test compound is also prepared for control.
  • the binding between TCTP and the binding partner can be quantified using SDS-PAGE or autoradiography.
  • Cytotoxic effects of a test compound can be determined using the following procedures.
  • Various tumor cell lines e.g., K562, U937, MDA-DB231, BT20, MCF7 can be used.
  • the cells are seeded at low density and are left for about 24 hours to allow recovery and reach a new logarithmic phase of growth.
  • Various concentrations of the compound being tested are added in triplicate. The mixture is incubated for about 6 days, during which approximately 4 doublings of the cell population occur, to allow the cells to reach subconfluence.
  • the viability of the cultured cells after the treatment by the test compound is determined by quantifying the level of ATP, since the level of ATP is directly proportional to the number of viable cells present in the culture.
  • the ATP level can be quantified using, for example, CellTiter-Glo ® (Promega) luminescent test for cell viability.
  • the rate of apoptosis can be assessed by using Poly ADP-Ribose Polymerase (PARP) cleavage test.
  • Tumor cells e.g., U937 cells
  • the proteins are isolated from these cells and loaded onto a gel for a western blot analysis.
  • a specific anti-PARP antibody is used to visualize the location of PARP. Cleavage of PARP indicates the induction of cell apoptosis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Neurosurgery (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biotechnology (AREA)
  • Toxicology (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

This invention encompasses proteic binding partners of Translationally Controlled Tumor Protein (TCTP), which can modulate tumor reversion or cell apoptosis. Methods of screening for compounds useful for treating, preventing or managing cancer or a neurodegenerative disorder and use of these compounds are also encompassed.

Description

PROTEIC BINDING PARTNERS OF TCTP AND METHODS OF MODULATING TUMOR REVERSION OR CELL APOPTOSIS
This application claims the benefit of U.S. Provisional Application No. 60/441,770, filed January 23, 2003, the entirety of which is incorporated herein by reference.
1. FIELD OF THE INVENTION
This invention encompasses methods of modulating tumor reversion or cell apoptosis. Also encompassed are methods of identifying compounds useful for treating, preventing and managing cancer or a neurodegenerative disorder.
2. BACKGROUND OF THE INVENTION
Cancer is characterized primarily by an increase in the number of abnormal cells derived from a given normal tissue, invasion of adjacent tissues by these abnormal cells, or lymphatic or blood-borne spread of malignant cells to regional lymph nodes and to distant sites (metastasis). Clinical data and molecular biologic studies indicate that cancer is a multi-step that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia. The neoplastic region may evolve clonically and develop an increasing capacity for invasion, growth, metastasis, and heterogenicity, especially under conditions in which neoplastic cells escape the host's immune surveillance. Roitt et al., Immunology, 17.1-17.12 (3rd Ed., Mosby, St. Louis, 1993).
Tumor reversion is a process wherein malignant cells revert to more normal phenotypes. The study of tumor reversion has identified genes that are differentially expressed between tumor cells and their revertants. Tuynder et al., Proc. Natl. Acad. Sci. USA, 99(23): 14976-14981 (2002). One of those is the gene tptl, which produces the Translationally Controlled Tumor Protein ("TCTP"), which has been found to be overexpressed in tumor cells, but not in the revertants. Id. On the other hand, neurodegenerative disorders are characterized by progressive and irreversible loss of neurons from specific regions of the brain. As a group, these disorders are relatively common and represent a substantial medical and social problem. They are primarily disorders of later life, developing in individuals who are neurologically normal, although childhood-onset forms of each of the disorders are recognized. Certain genes that are activated or suppressed during the cascade leading to tumor reversion or cell apoptosis have been identified and disclosed in, for example, WO 97/22695 and WO 00/08147, both of which are incorporated herein in their entirety by reference. One of the genes that is suppressed in the cascade, "TSLP2," was later found to be identical to presenilin 1, whose role had been implicated with Alzheimer's disease. Therefore, compounds that modulate tumor reversion or cell apoptosis present a potential venue in searching for an effective treatment, prevention and management of cancer or a neurodegenerative disorder.
3. SUMMARY OF THE INVENTION
This invention is based, in part, on the inventors' discovery that certain proteins, i.e., proteic binding partners of TCTP, interact with TCTP, and modulate (i.e., enhance/promote or reduce/inhibit) tumor reversion and/or cell apoptosis. Accordingly, this invention encompasses a method of modulating tumor reversion and/or cell apoptosis comprising contacting a tumor cell with an effective amount of a proteic binding partner of TCTP.
This invention also encompasses a method of identifying a compound that reduces or inhibits the binding between TCTP and a proteic binding partner of TCTP. Compounds so identified can be further screened for their efficacy in promoting or reducing/inhibiting tumor reversion or cell apoptosis.
This invention further encompasses methods of treating, preventing or managing cancer or a neurodegenerative disorder using the compounds identified using screening methods of this invention. In some embodiments, proteic binding partners of TCTP can be directly used for the treatment, prevention or management. Some proteic binding partners of TCTP, which promotes tumor reversion or cell apoptosis, can be used for the treatment, prevention or management of cancer, while other proteic binding partners of TCTP, which reduces or inhibits tumor reversion or cell apoptosis, can be used for the treatment, prevention or management of a neurodegenerative disorder. In other embodiments, other compounds, selected using methods based on the interaction between TCTP and its proteic binding partners, are used for the treatment, prevention or management of cancer or a neurodegenerative disorder. 4. BRIEF DESCRIPTION OF FIGURES
Figure 1 illustrates the inhibition of GTP-GDP exchange activity of Arfl- GAP by TCTP.
5. DETAILED DESCRIPTION OF THE INVENTION
TCTP is a ubiquitously expressed protein that is evolutionarily conserved. Its role in tumor reversion and/or cell apoptosis has been implicated. Tuynder et al., Proc. Natl. Acad. Sci. USA, 99(23): 14976-81 (2002). This invention is based, in part, on the inventors' discovery that certain proteins, i.e., proteic binding partners of TCTP, can interact with TCTP, and thus may be used to modulate TCTP's effect on tumor reversion and/or cell apoptosis.
Accordingly, a first embodiment of this invention encompasses methods of modulating tumor reversion and/or cell apoptosis comprising contacting a tumor cell with an effective amount of a proteic binding partner of TCTP. As used herein, and unless otherwise specified, the terms "modulate,"
"modulating," and "modulation," when used in connection with tumor reversion or cell apoptosis, mean that the extent or intensity of such tumor reversion or cell apoptosis is altered or adjusted. The terms encompass increase, promotion, decrease, reduction and inhibition of tumor reversion or cell apoptosis. As used herein, or unless otherwise specified, the terms "promote,"
"promotion," and "increase," when used in connection with tumor reversion or cell apoptosis, mean that tumor reversion or cell apoptosis in a cell, treated using methods of this invention, is higher than the same type of cell without the treatment. In some embodiments, tumor reversion or cell apoptosis in a cell treated using methods of this invention is higher than that in the same type of cell without the treatment by about 10 percent, 30 percent, 50 percent, 70 percent, 100 percent, 200 percent or more.
As used herein, and unless otherwise specified, the terms "decrease," "reduce," and "reduction," when used in connection with tumor reversion or cell apoptosis, means that tumor reversion or cell apoptosis in a cell, treated using methods of this invention, is lower than the same type of cell without the treatment. In some embodiments, tumor reversion or cell apoptosis in a cell treated using methods of this invention is lower than that in the same type of cell without the treatment by about 10 percent, 30 percent, 50 percent, 70 percent or 90 percent or more. In this regard, the terms "inhibit" and "inhibition" mean that tumor reversion or cell apoptosis in a cell treated using methods of this invention is substantially abolished. In some embodiments, tumor reversion or cell apoptosis is inhibited where no or little such tumor reversion or cell apoptosis can be detected using a well-known analytical method such as SDS-PAGE, PARP cleavage, spectrophotometry, autoradiography, or chromatographic techniques.
As used herein, and unless otherwise specified, the term "proteic binding partner of TCTP" means a protein, or portions thereof, that can bind to or interact with TCTP. Any known methods of identification of a binding partner may be used, e.g., co- precipitation and fluorescence resonance energy transfer. A specific method is yeast two- hybrid system, derived from the system described by Finley and Brent (Interaction trap cloning with yeast, in DNA Cloning, Expression Systems: A Practical Approach, pp. 169-203, Oxford Universal Press, Oxford (1995), which is incorporated herein by reference). Examples of proteic binding partners of TCTP include, but are not limited to, human elongation factor- 1 Delta and those listed in Table 1.
5.1 Methods of Screening
In one embodiment, this invention encompasses methods of identifying compounds that reduce or inhibit binding between TCTP and a proteic binding partner of TCTP comprising: (a) contacting a compound with TCTP and the proteic binding partner of TCTP; and (b) determining whether the binding between TCTP and the proteic binding partner of TCTP is decreased.
As used herein, and unless otherwise indicated, the terms "reduce" and "decrease," when used in connection with binding, mean that the binding between two molecules are impeded, slowed or prevented in the presence of a test compound as compared to the binding occurring in the absence of such a test compound. Binding between two molecules may be determined by any methods known in the art, including, but not limited to, fluorescence, spectroscopy, chromatography, centrifugation and electrophoresis. In some embodiments, for a test compound to be identified positive, binding between TCTP and a proteic binding partner of TCTP is reduced by about 10 percent, 20 percent, 40 percent, 60 percent, 80 percent or 90 percent or more in the presence of the test compound as compared to the binding of the same molecules in the absence of the test compound. In another embodiment, this invention encompasses methods of identifying compounds that are useful in treating, preventing or managing cancer. One aspect of this embodiment encompasses methods of identifying compounds that promote tumor reversion or cell apoptosis comprising: (a) contacting a proteic binding partner of TCTP with a tumor cell; and (b) determining whether tumor reversion or cell apoptosis is increased as compared to a tumor cell that has not been contacted with the proteic binding partner of TCTP. The proteic binding partner identified positive using the methods can be used in treating, preventing or managing cancer.
In another aspect of this embodiment, this invention also encompasses methods of identifying compounds that promote tumor reversion or cell apoptosis comprising: (a) contacting a compound with TCTP and a proteic binding partner of TCTP; (b) determining whether binding between TCTP and the proteic binding partner of TCTP is decreased; and wherein the binding between TCTP and the proteic binding partner of TCTP is decreased, then (c) contacting the compound with a tumor cell; and (d) determining whether tumor reversion or cell apoptosis is increased as compared to a tumor cell that has not been contacted with the compound. Compounds tested positive using this method is potentially useful for the treatment, prevention and/or management of cancer.
Conversely, this invention also encompasses methods of identifying compounds potentially useful in the treatment, prevention and/or management of a neurodegenerative disorder such as Alzheimer's disease. One aspect of this embodiment encompasses methods of identifying compounds that reduce or inhibit tumor reversion or cell apoptosis comprising: (a) contacting a proteic binding partner of TCTP with a tumor cell; and (b) determining whether tumor reversion or cell apoptosis is reduced as compared to a tumor cell that has not been contacted with the proteic binding partner of TCTP.
In another aspect of this embodiment, this invention also encompasses methods of identifying compounds that reduce or inhibit tumor reversion or cell apoptosis comprising: (a) contacting a compound with TCTP and a proteic binding partner of TCTP; (b) determining whether binding between TCTP and the proteic binding partner of TCTP is decreased; and wherein the binding between TCTP and the proteic binding partner of TCTP is decreased, then (c) contacting the compound with a tumor cell; and (d) determining whether tumor reversion or cell apoptosis is reduced as compared to a tumor cell that has not been contacted with the compound. Whether tumor reversion or cell apoptosis increases or decreases in the tumor cell contacted with the test compound can be determined by any methods known in the art, as well as those described herein. The determination can be done in vivo or in vitro. For example, where the tests are carried out in vitro, models such as K256/KS cells described in Telerman et al., Proc. Natl. Acad. Sci. USA, 90: 8702-6 (1993), Ml-LTR cells described in Amson et al., Proc. Natl. Acad. Sci. USA, 93: 3953-7 (1996), and U937/US3-US4 cells described in Nemani et al., Proc. Natl. Acad. Sci. USA, 93: 9039-42 (1996), all of which are incorporated herein by reference, may be used. In the case of in vivo tests, the test compound can be injected into an animal model, such as immunodepressed mice, and effects of the injected test compound may be investigated. Specific conditions and thresholds for identification are well within the ordinary skill in the art.
In the case of cell apoptosis, a specific method that can be used is Poly ADP-Ribose Polymerase (PARP) cleavage test. In short, tumor cells are treated with a test compound, and proteins are isolated. An anti-PARP can be used to visualize the location of PARP, which, in turn, provides an indication of whether PARP cleavage has occurred or not. Cleavage of PARP is an indication of the induction of cell apoptosis.
5.2 Methods of Treatment Prevention and Management In one embodiment, this invention encompasses methods of treating, preventing and/or managing cancer comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of a compound identified using certain methods of this invention.
As used herein, and unless otherwise specified, the terms "treat," "treating " and "treatment" refer to the eradication or amelioration of a disease or condition, or of one or more symptoms associated with the disease or condition. In certain embodiments, the terms refer to minimizing the spread or worsening of the disease or condition resulting from the administration of one or more prophylactic or therapeutic agents to a subject with such a disease or condition. As used herein, and unless otherwise specified, the terms "prevent,"
"preventing" and "prevention" refer to the prevention of the onset, recurrence or spread of a disease or condition, or of a symptom thereof.
As used herein, and unless otherwise specified, the terms "manage," "managing" and "management" refer to preventing or slowing the progression, spread or worsening of a disease or condition. Often, the beneficial effects that a subject derives from a prophylactic or therapeutic agent do not result in a cure of the disease or condition. As used herein and unless otherwise indicated, the term "managing" encompasses preventing the recurrence of cancer in a patient who had suffered from cancer, lengthening the time a patient who had suffered from cancer remains in remission, preventing the occurrence of cancer in patients at risk of suffering from cancer (e.g., patients who had been exposed to high amounts of radiation or carcinogenic materials, such as asbestos; patients infected with viruses associated with the occurrence of cancer, such as, but not limited to, HIV and Kaposi's sarcoma-associated herpes virus; and patients with genetic predispositions to cancer, such as those suffering from Downs syndrome), and preventing the occurrence of malignant cancer in patients suffering from pre-malignant or non-malignant cancers.
As used herein, and unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment or management of a disease or condition, or to delay or minimize one or more symptoms associated with the disease or condition. A therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other therapies, which provides a therapeutic benefit in the treatment or management of the disease or condition. The term "therapeutically effective amount" can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease or condition, or enhances the therapeutic efficacy of another therapeutic agent.
As used herein, and unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease or condition, or prevent its recurrence. A prophylactically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other agents, which provides a prophylactic benefit in the prevention of the disease. The term "prophylactically effective amount" can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent. As used herein, the term "cancer" includes, but is not limited to, solid tumors and blood born tumors. The term "cancer" refers to disease of skin tissues, organs, blood, and vessels, including, but not limited to, cancers of the bladder, bone or blood, brain, breast, cervix, chest, colon, endrometrium, esophagus, eye, head, kidney, liver, lymph nodes, lung, mouth, neck, ovaries, nancreas, prostate, rectum, stomach, testis, throat, and uterus. Specific cancers include, but are not limited to, advanced malignancy, amyloidosis, neuroblastoma, meningioma, hemangiopericytoma, multiple brain metastases, glioblastoma multiforms, glioblastoma, brain stem glioma, poor prognosis malignant brain tumor, malignant glioma, recurrent malignant giolma, anaplastic astrocytoma, anaplastic oligodendroglioma, neuroendocrine tumor, rectal adenocarcinoma, Dukes C & D colorectal cancer, unresectable colorectal carcinoma, metastatic hepatocellular carcinoma, Kaposi's sarcoma, karotype acute myeloblastic leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma, low grade follicular lymphoma, localized or metastatic melanoma (of any kind, including, but not limited to, ocular), peritoneal carcinoma, papillary serous carcinoma, gynecologic sarcoma, soft tissue sarcoma, scelroderma, cutaneous vasculitis, Langerhans cell histiocytosis, leiomyosarcoma, fϊbrodysplasia ossifϊcans progressive, hormone refractory prostate cancer, resected high-risk soft tissue sarcoma, unrescectable hepatocellular carcinoma, Waldenstrom's macroglobulinemia, smoldering myeloma, indolent myeloma, fallopian tube cancer, androgen independent prostate cancer, androgen dependent stage IV non-metastatic prostate cancer, hormone-insensitive prostate cancer, chemotherapy- insensitive prostate cancer, papillary thyroid carcinoma, follicular thyroid carcinoma, medullary thyroid carcinoma, and leiomyoma. In a specific embodiment, the cancer is metastatic. In another embodiment, the cancer is refractory or resistance to chemotherapy or radiation.
In some embodiments of this invention, compounds of this invention can be administered, sequentially or simultaneously, with another anticancer agent. The administration may be via same route or different routes. Examples of anti-cancer agents include, but are not limited to: acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; celecoxib; chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; Anvnπ ibicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; iproplatin; irinotecan; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; taxotere; tegafur; teloxantrone hydrochloride; temoporfϊn; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; and zorubicin hydrochloride.
Other anti-cancer drugs include, but are not limited to: 20-epi-l,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morpho genetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-aletbine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN
700; cartilage derived inliibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin
816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; dihydrotaxol, 9-; dioxamycin; diphenyl spiromustine; docetaxel; docosanol; dolasetron; doxifluridine; doxorubicin; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imatinib (e.g., Gleevec®), imiquimod; immunostimulant peptides; insulin-like growth factor- 1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inliibitor; mifepristone; miltefosine; mirimostim; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim;Erbitux, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; oblimersen (Genasense®); O6"benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetm A; placetin B; plasminogen activator inhibitor; platinum complex; platinum compounds; platinum-triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RTI retinamide; rohitukine; romurtide; roquinimex; rubiginone Bl; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction inhibitors; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; tallimustine; tamoxifen metl iodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.
The magnitude of a prophylactic or therapeutic dose of each active ingredient in the treatment, prevention and/or management of cancer will typically vary with the specific active ingredients, the severity and type of cancer, and the route of administration. The dose, and perhaps the dose frequency, may also vary according to age, body weight, response, and the past medical history of the patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors by following, for example, dosages reported in the literature and recommended in the Physician 's Desk Reference® (54th ed., 2000).
In one embodiment, the patient is an animal. In another embodiment, the patient is a mammal, specifically, a human.
In another embodiment, this invention encompasses methods of treating, preventing and managing a neurodegenerative disorder comprising administering to a patient in need of such treatment, prevention and/or management a therapeutically or prophylactically effective amount of a compound identified certain methods of this invention.
Examples of neurodegenerative disorders that can be treated using methods of this invention include, but are not limited to, amylotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease and Parkinson's disease. In one embodiment, the neurodegenerative disorder that can be treated, prevented and/or managed using methods of this invention is Alzheimer's disease, hi one embodiment, the patient is a mammal, in particular, a human. In some embodiments, compound of this invention may be administered, sequentially or simultaneously, with another agent useful in treating a neurodegenerative disorder. The administration may be via same route or different routes. Examples of other agents include, but are not limited to: acridine derivatives such as tacrine; amantadine; dopamine depleting agents such as tetrabenazine and reserpine; dopamine receptor antagonists such as bromocriptine and pergolide; metabolic precursors of dopamine such as levodopa; inhibitors of acetylcholinesterase (AChE) such as physostigmine; inhibitors of L-amino acid decarboxylase such as benserazide and carbidopa; muscarinic receptor antagonists such as benzotropine mesylate, diphenhydramine hydorchloride and trihexyphenidyl; precursors of acetylcholine synthesis such as choline chloride and phosphatidyl choline (lecithin); and selegiline.
The magnitude of a prophylactic or therapeutic dose of each active ingredient in the treatment, prevention and/or management of a neurodegenerative disorder will typically vary with the specific active ingredients, the severity and type of disorder, and the route of administration. The dose, and perhaps the dose frequency, may also vary according to age, body weight, response, and the past medical history of the patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors by following, for example, dosages reported in the literature and recommended in the Physician 's Desk Reference® (54m ed., 2000).
5.3 Pharmaceutical Compositions
In some embodiments, this invention encompasses pharmaceutical compositions comprising one or more proteic binding partners of TCTP, or genes thereof.
In other embodiments, this invention also encompasses pharmaceutical compositions comprising one or more of compounds identified using methods of this invention. Such pharmaceutical compositions are described below.
Certain pharmaceutical compositions are single unit dosage forms suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial), or transdermal administration to a patient. Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols ( e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
The formulation should suit the mode of administration. For example, oral administration requires enteric coatings to protect the compounds of this invention from degradation within the gastrointestinal tract. In another example, the compounds of this invention may be administered in a liposomal formulation to shield the compounds from degradative enzymes, facilitate transport in circulatory system, and effect delivery across cell membranes to intracellular sites.
The composition, shape, and type of dosage forms of the invention will typically vary depending on their use. For example, a dosage form used in the acute treatment of a disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease. Similarly, a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease. These and other ways in which specific dosage forms encompassed by this invention will vary from one another will be readily apparent to those skilled in the art. See, e.g., Remington 's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
5.3.1 Oral Dosage Forms Pharmaceutical compositions of the invention that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups). Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
Typical oral dosage forms of the invention are prepared by combining the active ingredients in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
Disintegrants or lubricants can be used in pharmaceutical compositions and dosage forms of the invention.
5.3.2 Parenteral Dosage Forms
Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, 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 such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl rnyristate, and benzyl benzoate.
Compounds that increase the solubility of one or more of the active ingredients (i.e., the compounds of this invention) disclosed herein can also be incorporated into the parenteral dosage forms of the invention.
5.3.3 Transdermal, Topical and Mucosal Dosage Forms
Transdermal, topical, and mucosal dosage forms of the invention include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington 's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980 & 1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985). Transdermal dosage forms include "reservoir type" or "matrix type" patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
Suitable excipients (e.g., carriers and diluents) and other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
Depending on the specific tissue to be treated, additional components may be used prior to, in conjunction with, or subsequent to treatment with active ingredients of the invention. For example, penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
The pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied, may also be adjusted to improve delivery of one or more active ingredients. Similarly, the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery. Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery. In this regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent. Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
5.3.4 Compositions with Enhanced Stability
The suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water. Active ingredients that comprise primary or secondary amines are particularly susceptible to such accelerated decomposition. Consequently, this invention encompasses pharmaceutical compositions and dosage forms that contain little, if any, lactose other mono- or di-saccharides. As used herein, the term "lactose-free" means that the amount of lactose present, if any, is insufficient to substantially increase the degradation rate of an active ingredient.
Lactose- free compositions of the invention can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) 25-NF20 (2002). In general, lactose-free compositions comprise active ingredients, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts. Preferred lactose-free dosage forms comprise active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate.
This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water (e.g., 5%) is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80. In effect, water and heat accelerate the decomposition of some compounds. Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations. Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anliydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected. An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are preferably packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
The invention further encompasses pharmaceutical compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as "stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
Like the amounts and types of excipients, the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients.
5.3.5 Delayed Release Dosage Forms
Active ingredients of the invention can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is incorporated herein by reference. Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microp articles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the compounds of this invention. The invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
All controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts. Ideally, the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time. Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance. In addition, controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
Most controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
5.3.6 Delivery of Nucleic Acids
Active ingredients in pharmaceutical compositions of this invention may be in the form of nucleic acids. Delivery of these molecules into a patient can either be direct, i.e., the patient is directly exposed to the compounds of this invention or compound-carrying vector, or indirect, i.e., cells are first transformed with the compounds of this invention in vitro, then transplanted into the patient for cell replacement therapy. These two approaches are known as in vivo and ex vivo therapy, respectively. In the case of in vivo therapy, compounds of this invention are directly administered in vivo, where they are expressed to produce the encoded product. This can be accomplished by any of numerous methods known in the art, e.g., by constructing them as part of an appropriate nucleic acid expression vector and administering them so that they become intracellular, by infection using a defective or attenuated retroviral or other viral vector (see, e.g., U.S. Patent No. 4,980,286), by direct injection of naked DNA, by use of microparticle bombardment (e.g., a gene gun; Biolistic®, DuPont), by coating with lipids or cell-surface receptors or transfecting agents, encapsulation in liposomes, microparticles, or microcapsules, by administering them in linkage to a peptide which is known to enter the cell or nucleus, or by administering them in linkage to a ligand subject to receptor-mediated endocytosis (Wu and Wu, J Biol. Chem., 262: 4429-32 (1987)), which can be used to target cell types specifically expressing the receptors. Further, compounds of this invention can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor, as disclosed in, for example, WO 92/06180, WO 92/22635, WO 92/20316, WO 93/14188 and WO 93/20221, all of which are incorporated herein by reference.
Ex vivo therapy involves transferring the compounds of this invention to cells in tissue culture by methods such as electroporation, lipofection, calcium phosphate mediated transfection and viral infection. Usually, the method of transfer includes the transfer of a selectable marker to the cells. The r.ells are then placed under selection to isolate those cells that have taken up and are expressing the transferred compounds. These cells are then delivered to a patient.
Compounds of this invention are introduced into a cell prior to administration in vivo of the resulting recombinant cell. Such introduction can be carried out by any methods known in the art, including, but not limited to, transfection, electroporation, microinjection, infection with a viral vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer and spheroplast fusion. Numerous techniques are known in the art for the introduction of foreign compounds into cells. Examples of such techniques are disclosed in: Loeffler et al, Meth. Enzymol, 217: 599-618 (1993); Cohen et al, Meth. Enzymol, 217: 618-644 (1993); and Cline, Pharmc. Tlier., 29: 69-92 (1985), all of which are incorporated herein by reference. These techniques should provide for the stable transfer of the compounds of this invention to the cell, so that they are expressible by the cell and preferably heritable and expressible by its cell progeny. The resulting recombinant cells can be delivered to a patient by various methods known in the art. Examples of the delivery methods include, but are not limited to, subcutaneous injection, skin graft and intravenous injection.
5.3.7 Kits In some cases, active ingredients of the invention are preferably not administered to a patient at the same time or by the same route of administration. This invention therefore encompasses kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a patient.
A typical kit of the invention comprises a single unit dosage form of the compounds of this invention, or a pharmaceutically acceptable salt, prodrug, solvate, hydrate, clathrate or stereoisomer thereof, and a single unit dosage form of another agent that may be used in combination with the compounds of this invention. Eats of the invention can further comprise devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, drip bags, patches, and inhalers.
Kits of the invention can further comprise pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, 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 such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
The invention is further defined by reference to the following non-limiting examples. It will be apparent to those skilled in the art that many modifications, both to materials and methods, can be practiced without departing from the spirit and scope of this invention.
6. EXAMPLES 6.1 Identification of Proteic Binding Partners of TCTP
Two separate yeast two hybrid hunts were performed with TCTP. One involved using the full-length TCTP as bait, and the second involved using the first 80 amino acids of TCTP as bait. A cDNA library derived from U937 cell line was used to identify the proteic binding partners of TCTP. When the full-length TCTP was used, human elongation factor- 1 delta
(Accession No. NM_032378; NM_001960) was identified as the binding partner of TCTP. The results from the two hybrid hunt using the first 80 amino acids of TCTP are shown below in Table 1.
Table 1. Proteic Binding Partners of TCTP
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
6.2 Inhibition of GTP-GDP Exchange Activity of Arfl-GAP by TCTP
Into a buffer containing 1 mM free Mg2+, 30 uM TCTP and 0.5 uM [D17JARF1-GDP were added. The reaction was initiated by the addition of 40 uM GTP and 2 mM EDTA. To stop the reaction, 2 mM MgCl2 was added. The Arf-GAP, at a concentration of 100 uM, was added. The reaction was measured in presence and absence of TCTP. The tryptophan fluorescence change that accompanies the conformational change of ARF1 from the inactive state (GDP -bound) to active state (GTP -bound) was followed in real-time to assess the activation of [D17JARF1. As shown in Figure 1 , the results show that the function of Arfl , a binding partner of TCTP, is affected by the presence of TCTP. In particular, it is shown that TCTP inhibits the GTP-GDP exchange catalyzed by Arfl -GAP, implicating that Arfl is kept in its active conformation in presence of TCTP.
6.3 Identification of a Compound that Reduces or Inhibits
Binding between TCTP and its Binding Partner
Radio-labeled TCTP is generated from an in vitro translation in a rabbit reticulocyte lysate in the presence of 35S-labeled methionine and 35S-labeled cysteine. A
GST-fusion protein of a binding partner of TCTP is prepared, and is coupled to sepharose-glutathione beads. 35S labeled TCTP, sepharose-glutathione coupled binding partner and a test compound are mixed and incubated for a sufficient time. A mixture without the test compound is also prepared for control. The binding between TCTP and the binding partner can be quantified using SDS-PAGE or autoradiography.
6.4 Cytotoxic Effects of a Test Compound
One indicator of an anti-cancer agent is its cytotoxic activity. Cytotoxic effects of a test compound can be determined using the following procedures.
Various tumor cell lines (e.g., K562, U937, MDA-DB231, BT20, MCF7) can be used. The cells are seeded at low density and are left for about 24 hours to allow recovery and reach a new logarithmic phase of growth. Various concentrations of the compound being tested are added in triplicate. The mixture is incubated for about 6 days, during which approximately 4 doublings of the cell population occur, to allow the cells to reach subconfluence.
The viability of the cultured cells after the treatment by the test compound is determined by quantifying the level of ATP, since the level of ATP is directly proportional to the number of viable cells present in the culture. The ATP level can be quantified using, for example, CellTiter-Glo® (Promega) luminescent test for cell viability.
6.5 Effects on Cell Apoptosis
The rate of apoptosis can be assessed by using Poly ADP-Ribose Polymerase (PARP) cleavage test. Tumor cells (e.g., U937 cells) are treated with various concentrations of a test compound for 24 hours. The proteins are isolated from these cells and loaded onto a gel for a western blot analysis. A specific anti-PARP antibody is used to visualize the location of PARP. Cleavage of PARP indicates the induction of cell apoptosis.
All of the patents, patent applications and publications referred to in this application are incorporated herein in their entirety by reference. Moreover, citation or identification of any reference in this application is not an admission that such reference is available as prior art to this invention. The full scope of the invention is better understood with reference to the appended claims.

Claims

CLAIMSWhat is claimed is:
1. A method of modulating tumor reversion or cell apoptosis comprising contacting a tumor cell with an effective amount of a proteic binding partner of TCTP.
2. The method of claim 1 , wherein the proteic binding partner of TCTP is one of the following:
Figure imgf000028_0001
Figure imgf000029_0001
or a combination thereof.
3. A method of identifying a compound that reduces or inhibits binding between TCTP and a proteic binding partner of TCTP comprising:
(a) contacting the compound with TCTP and the proteic binding partner of TCTP; and
(b) determining whether the binding between TCTP and the proteic binding partner of TCTP is decreased.
4. The method of claim 3, wherein the proteic binding partner of TCTP is:
Figure imgf000030_0001
Figure imgf000031_0001
5. A method of identifying a compound that promotes tumor reversion or cell apoptosis comprising:
(a) contacting a proteic binding partner of TCTP with a tumor cell; and
(b) determining whether tumor reversion or cell apoptosis is increased as compared to a tumor cell that has not been contacted with the proteic binding partner of TCTP.
6. A method of identifying a compound that promotes tumor reversion or cell apoptosis comprising: (a) contacting the compound with TCTP and a proteic binding partner of TCTP; (b) determining whether binding between TCTP and the proteic binding partner of TCTP is decreased; and wherein the binding between TCTP and the proteic binding partner is decreased, then (c) contacting the compound with a tumor cell; and
(d) determining whether tumor reversion or cell apoptosis is increased as compared to a tumor cell that has not been contacted with the compound.
7. The method of claim 5 or 6, wherein the proteic binding partner of TCTP is:
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
8. A method of treating, preventing or managing cancer comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of the compound identified using the method of claim 5 or 6.
9. A method of identifying a compound that reduces or inhibits tumor reversion or cell apoptosis comprising:
(a) contacting a proteic binding partner of TCTP with a tumor cell; and
(b) determining whether tumor reversion or cell apoptosis is reduced as compared to a tumor cell that has not been contacted with the proteic binding partner of TCTP.
10. A method of identifying a compound that reduces or inhibits tumor reversion or cell apoptosis comprising:
(a) contacting the compound with TCTP and a proteic binding partner of TCTP;
(b) determining whether binding between TCTP and the proteic binding partner of TCTP is decreased; and wherein the binding between TCTP and the proteic binding partner is decreased, then
(c) contacting the compound with a tumor cell; and (d) determining whether tumor reversion or cell apoptosis is reduced as compared to a tumor cell that has not been contacted with the compound.
11. The method of claim 9 or 10, wherein the proteic binding partner of TCTP
Figure imgf000035_0001
Figure imgf000036_0001
12. A method of treating, preventing or managing a neurodegenerative disorder comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of the compound identified using the method of claim 9 or 10.
13. The method of claim 12, wherein the neurodegenerative disorder is Alzheimer's disease.
PCT/US2004/001894 2003-01-23 2004-01-23 Proteic binding partners of tctp and methods of modulating tumor reversion or cell apoptosis WO2004065572A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/187,891 US20060140970A1 (en) 2003-01-23 2005-07-25 Proteic binding partners of TCTP and methods of modulating tumor reversion or cell apoptosis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US44177003P 2003-01-23 2003-01-23
US60/441,770 2003-01-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/187,891 Continuation-In-Part US20060140970A1 (en) 2003-01-23 2005-07-25 Proteic binding partners of TCTP and methods of modulating tumor reversion or cell apoptosis

Publications (2)

Publication Number Publication Date
WO2004065572A2 true WO2004065572A2 (en) 2004-08-05
WO2004065572A3 WO2004065572A3 (en) 2004-11-04

Family

ID=32771971

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/001894 WO2004065572A2 (en) 2003-01-23 2004-01-23 Proteic binding partners of tctp and methods of modulating tumor reversion or cell apoptosis

Country Status (1)

Country Link
WO (1) WO2004065572A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012080509A1 (en) * 2010-12-17 2012-06-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nucleic acids targeting tctp for use in the treatment of chemo-or hormone-resistant cancers
US8435959B2 (en) 2004-03-29 2013-05-07 University Of South Florida Effective treatment of tumors and cancer with triciribine and related compounds
WO2013176623A1 (en) * 2012-05-21 2013-11-28 Agency For Science, Technology And Research (A*Star) Inhibitors for the treatment of cancer
US8623834B2 (en) 2004-03-29 2014-01-07 University Of South Florida Compositions including triciribine and trastuzumab and methods of use thereof
US8673868B2 (en) 2004-03-29 2014-03-18 University Of South Florida Compositions including triciribine and one or more platinum compounds and methods of use thereof
US8673867B2 (en) 2004-03-29 2014-03-18 University Of South Florida Compositions including triciribine and epidermal growth factor receptor inhibitor compounds or salts thereof and methods of use thereof
US8691779B2 (en) 2004-03-29 2014-04-08 University Of South Florida Compositions including triciribine and taxanes and methods of use thereof
US8865666B2 (en) 2004-03-29 2014-10-21 University Of South Florida Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
CN111228292A (en) * 2020-03-11 2020-06-05 福州载基生物科技有限公司 Application of human TPT1/TCTP gene in preparation of antitumor drugs

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994012881A2 (en) * 1992-12-02 1994-06-09 Hochstrasser Denis F A METHOD FOR DETECTING GROWING CELLS USING TRANSLATIONALLY CONTROLLED TUMOR PROTEIN p21
WO2002036624A2 (en) * 2000-10-30 2002-05-10 Board Of Regents, The University Of Texas System Methods, compositions and therapeutic applications relating to fortilin (= translationally controlled tumor protein, tctp) and modulators thereof
US20020177692A1 (en) * 2001-04-16 2002-11-28 Myriad Genetics, Incorporated BCL-XL-interacting protein and use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994012881A2 (en) * 1992-12-02 1994-06-09 Hochstrasser Denis F A METHOD FOR DETECTING GROWING CELLS USING TRANSLATIONALLY CONTROLLED TUMOR PROTEIN p21
WO2002036624A2 (en) * 2000-10-30 2002-05-10 Board Of Regents, The University Of Texas System Methods, compositions and therapeutic applications relating to fortilin (= translationally controlled tumor protein, tctp) and modulators thereof
US20030172388A1 (en) * 2000-10-30 2003-09-11 Ken Fujise Methods and compositions relating to fortilin, an anti-apoptotic molecule, and modulators of fortilin
US20020177692A1 (en) * 2001-04-16 2002-11-28 Myriad Genetics, Incorporated BCL-XL-interacting protein and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
TUYNDER M ET AL: "Biological models and genes of tumor reversion: cellular reprogramming through tpt1/TCTP and SIAH-1" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 99, no. 23, 12 November 2002 (2002-11-12), pages 14976-14981, XP002226444 ISSN: 0027-8424 *

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9186403B2 (en) 2004-03-29 2015-11-17 University Of South Florida Compositions including triciribine and trastuzumab and methods of use thereof
US8673868B2 (en) 2004-03-29 2014-03-18 University Of South Florida Compositions including triciribine and one or more platinum compounds and methods of use thereof
US9655914B2 (en) 2004-03-29 2017-05-23 University Of South Florida Compositions including triciribine and epidermal growth factor receptor inhibitor compounds or salts thereof and methods of use thereof
US9211299B2 (en) 2004-03-29 2015-12-15 University Of South Florida Compositions including triciribine and one or more platinum compounds and methods of use thereof
US9192645B2 (en) 2004-03-29 2015-11-24 University Of South Florida Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
US8673867B2 (en) 2004-03-29 2014-03-18 University Of South Florida Compositions including triciribine and epidermal growth factor receptor inhibitor compounds or salts thereof and methods of use thereof
US8691779B2 (en) 2004-03-29 2014-04-08 University Of South Florida Compositions including triciribine and taxanes and methods of use thereof
US8865666B2 (en) 2004-03-29 2014-10-21 University Of South Florida Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
US9101641B2 (en) 2004-03-29 2015-08-11 University Of South Florida Compositions including triciribine and taxanes and methods of use thereof
US9115162B2 (en) 2004-03-29 2015-08-25 University Of South Florida Effective treatment of tumors and cancer with triciribine and related compounds
US9518079B2 (en) 2004-03-29 2016-12-13 University Of South Florida Effective treatment of tumors and cancer with triciribine and related compounds
US8435959B2 (en) 2004-03-29 2013-05-07 University Of South Florida Effective treatment of tumors and cancer with triciribine and related compounds
US8623834B2 (en) 2004-03-29 2014-01-07 University Of South Florida Compositions including triciribine and trastuzumab and methods of use thereof
US9265783B2 (en) 2004-03-29 2016-02-23 University Of South Florida Compositions including triciribine and epidermal growth factor receptor inhibitor compounds or salts thereof and methods of use thereof
US9486492B2 (en) 2004-03-29 2016-11-08 University Of South Florida Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
US9486522B2 (en) 2004-03-29 2016-11-08 University Of South Florida Compositions including triciribine and trastuzumab and methods of use thereof
US9486470B2 (en) 2004-03-29 2016-11-08 University Of South Florida Compositions including triciribine and one or more platinum compounds and methods of use thereof
US9498492B2 (en) 2004-03-29 2016-11-22 University Of South Florida Compositions including triciribine and taxanes and methods of use thereof
WO2012080509A1 (en) * 2010-12-17 2012-06-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nucleic acids targeting tctp for use in the treatment of chemo-or hormone-resistant cancers
WO2013176623A1 (en) * 2012-05-21 2013-11-28 Agency For Science, Technology And Research (A*Star) Inhibitors for the treatment of cancer
CN111228292A (en) * 2020-03-11 2020-06-05 福州载基生物科技有限公司 Application of human TPT1/TCTP gene in preparation of antitumor drugs

Also Published As

Publication number Publication date
WO2004065572A3 (en) 2004-11-04

Similar Documents

Publication Publication Date Title
US20040072824A1 (en) Methods and compositions for the treatment of cancer
US10973822B2 (en) Combination therapy for treatment of hematological cancers and solid tumors
TWI509247B (en) Methods for the treatment of non-hodgkin's lymphomas using lenalidomide, and gene and protein biomarkers as a predictor
ES2940302T3 (en) Methods for treating multiple myeloma with immunomodulatory compounds in combination with antibodies
US20230310354A1 (en) Use of cyp26-resistant rar alpha selective agonists in the treatment of cancer
WO2012050374A2 (en) Immunotherapy for solid tumors
CN103142594A (en) Method using 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione for treatment of certain leukemias
CN102908346A (en) Use of 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione for treatment of mantle cell lymphomas
WO2003097835A2 (en) Pharmaceutical compositions for the treatment of cancer
US20060140970A1 (en) Proteic binding partners of TCTP and methods of modulating tumor reversion or cell apoptosis
EP2134341B1 (en) Method of treating cell proliferative disorders using growth hormone secretagogues
US10159675B2 (en) Cycling therapy using 3-(5-amino-2-methyl-4-oxo-4H-quinazolin-3-yl)-piperidine-2,6-dione
WO2004065572A2 (en) Proteic binding partners of tctp and methods of modulating tumor reversion or cell apoptosis
WO2016164336A1 (en) Hepatocellular carcinoma treatment using a combination of cc 122 and sorafenib
EP1656124A1 (en) Methods and compositions for the treatment of cancer
KR100898330B1 (en) Novel cytotoxic flavone derivative 7-O-3-benzyloxypropyl5,4' -di-O-methylapigenin, the preparation method and composition for treating cancers comprising the compound
KR20090061358A (en) Composition for preventing and treating breast cancer comprising acacetin
JP7473483B2 (en) Methods for treating cancer characterized by high expression levels of the spindle- and kinetochore-associated complex subunit 3 (SKA3) gene - Patents.com
WO2008036954A1 (en) Use of n- [5- [ [ [5- (1, 1-dimethylethyl) -2-oxazoyl] methyl] thio] -2-thiazolyl] -4-piperidinecarboxamide
US20210000845A1 (en) Methods of treating cancer in pediatric patients
WO2007143096A2 (en) Compounds for treating cancers
WO2008073304A9 (en) Cancer treatment methods
KR20090061356A (en) Composition for treating and preventing breast cancer comprising genistein

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 11187891

Country of ref document: US

122 Ep: pct application non-entry in european phase
WWP Wipo information: published in national office

Ref document number: 11187891

Country of ref document: US