US20070225350A1 - Compositions and methods for treating neoplastic diseases - Google Patents

Compositions and methods for treating neoplastic diseases Download PDF

Info

Publication number
US20070225350A1
US20070225350A1 US11/293,354 US29335405A US2007225350A1 US 20070225350 A1 US20070225350 A1 US 20070225350A1 US 29335405 A US29335405 A US 29335405A US 2007225350 A1 US2007225350 A1 US 2007225350A1
Authority
US
United States
Prior art keywords
cancer
compound
formula
cells
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/293,354
Other languages
English (en)
Inventor
Kenneth Anderson
Dharminder Chauhan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Celgene Research and Development I ULC
Dana Farber Cancer Institute Inc
Original Assignee
Dana Farber Cancer Institute Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dana Farber Cancer Institute Inc filed Critical Dana Farber Cancer Institute Inc
Priority to US11/293,354 priority Critical patent/US20070225350A1/en
Assigned to DANA-FARBER CANCER INSTITUTE reassignment DANA-FARBER CANCER INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANDERSON, KENNETH C., CHAUHAN, DHARMINDER
Publication of US20070225350A1 publication Critical patent/US20070225350A1/en
Priority to US12/183,007 priority patent/US8722724B2/en
Priority to US14/224,965 priority patent/US10610517B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: DANA-FARBER CANCER INST
Assigned to NATIONAL INSTITUTES OF HEALTH-DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH-DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: DANA-FARBER CANCER INSTITUTE
Assigned to TRIPHASE RESEARCH AND DEVELOPMENT I CORP. reassignment TRIPHASE RESEARCH AND DEVELOPMENT I CORP. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEREUS PHARMACEUTICALS, INC.
Priority to US16/800,570 priority patent/US20200206190A1/en
Priority to US17/890,563 priority patent/US20230210817A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to the fields of chemistry and medicine. More particularly, the present invention relates to the treatment of neoplastic diseases, such as cancer.
  • Cancer is a leading cause of death in the United States. Despite significant efforts to find new approaches for treating cancer, the primary treatment options remain surgery, chemotherapy and radiation therapy, either alone or in combination. Surgery and radiation therapy, however, are generally useful only for fairly defined types of cancer, and are of limited use for treating patients with disseminated disease. Chemotherapy is the method that is generally useful in treating patients with metastatic cancer or diffuse cancers such as leukemias. Although chemotherapy can provide a therapeutic benefit, it often fails to result in cure of the disease due to the patient's cancer cells becoming resistant to the chemotherapeutic agent.
  • the successful development of Bortezomib/PS-341 therapy for treatment of relapsed/refractory multiple myeloma (MM) has established proteasome inhibition as an effective therapeutic strategy.
  • the dipeptide boronic acid analogue Bortezomib is a potent, highly selective, and reversible proteasome inhibitor which targets the 26S proteasome complex and inhibits its function.
  • the 26S proteasome is an adenosine triphosphate (ATP)-dependent multicatalytic protease mediating intracellular protein degradation. Proteasomal degradation of misfolded or damaged proteins proceeds by recognition of polyubiquitinated proteins by the 19S regulatory subunit of the 26S protease, and subsequent hydrolysis to small polypeptides.
  • Bortezomib primarily inhibits chymotryptic, without altering tryptic or caspase-like, proteasome activity. Besides inhibiting NF-kB, Bortezomib has pleiotropic effects on MM biology by targeting: 1) cell-cycle regulatory proteins; 2) UPR pathway via modulating transcriptional activity of plasma cell differentiation factor X-box binding protein-1 (XBP-1); 3) p53-mediated apoptosis/MDM2; 4) DNA repair mechanisms; 5) classical stress-response pathways via both intrinsic (caspase-9 mediated) and extrinsic (caspase-8 mediated) cell death cascades.
  • XBP-1 plasma cell differentiation factor X-box binding protein-1
  • MDM2 p53-mediated apoptosis/MDM2
  • DNA repair mechanisms 5) classical stress-response pathways via both intrinsic (caspase-9 mediated) and extrinsic (caspase-8 mediated) cell death cascades.
  • Bortezomib activates JNK, which triggers mitochondrial apoptotic signaling: release of cytochrome-c (cyto-c) and second mitochondrial activator of caspases (Smac) from mitochondria to cytosol, followed by activation of caspase-9 and caspase-3.
  • cyto-c cytochrome-c
  • Smac second mitochondrial activator of caspases
  • One aspect of the present invention is a method of treating a neoplastic disease, comprising administering to a patient inflicted with the neoplastic disease a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof: wherein X is selected from the group consisting of fluorine, chlorine, bromine or iodine, and wherein the neoplastic disease is susceptible to resistance to at least one other chemotherapeutic agent.
  • Another aspect of the present invention is a method of treating a neoplastic disease, comprising administering to a patient inflicted with the neoplastic disease a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof wherein X is selected from the group consisting of fluorine, chlorine, bromine or iodine, in combination with at least one additional chemotherapeutic agent.
  • Another aspect of the present invention is a pharmaceutical composition, comprising a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof, wherein X is selected from the group consisting of fluorine, chlorine, bromine or iodine, and at least one additional chemotherapeutic agent.
  • Another aspect of the present invention is a method of treating a neoplastic disease, comprising administering to a patient inflicted with the neoplastic disease a synergistic combination of at least two proteosome inhibitors.
  • FIG. 1 illustrates inhibition of chymotrypsin-like, caspase-like, and trypsin-like proteasome activities in human erythrocytes-derived 20S proteasome by NPI-0052.
  • FIG. 2 illustrates the in vivo chymotrypsin-like activity of NPI-0052 in mice.
  • FIG. 3 depicts the autoradiograph obtained after treating MM.1S multiple myeloma (MM) cells with NPI-0052 (7 nM) and incubating protein extracts with AdaY( 125 I)Ahx 3 L 3 VS at 37° C.
  • FIG. 4 depicts immunoblots obtained after treating MM.1S cells with NPI-0052 and then incubating with Dansyl-Ahx3L3VS.
  • FIG. 5A illustrates cell viability of various multiple myeloma cell lines treated with indicated doses of NPI-0052 for 24 h.
  • FIG. 5B illustrates DNA fragmentation assays of apoptosis after treatment with NPI-0052 of MM cells obtained from patients.
  • FIG. 6 illustrates DNA fragmentation assays of apoptosis after treatment with NPI-0052 of bone marrow stromal cells obtained from patients.
  • FIG. 7 illustrates MTT assay of MM.1S cell viability after treatment with NPI-0052 or Dex in the presence or absence of IL-6 or IGF-I.
  • FIG. 8 illustrates the effect of NPI-0052 on VEGF-induced migration of MM.1S cells.
  • FIG. 9 illustrates the effect of NPI-0052 on Bcl2-overexpressing MM.1S cell viability.
  • FIGS. 10A and 10B depict the effect of NPI-0052 on tumor growth when administered orally to mice.
  • FIG. 10C illustrates the effect of NPI-0052 on survival when administered orally to mice.
  • FIG. 10D illustrates the effect of NPI-0052 on body weight when administered orally to mice.
  • FIG. 10E illustrates tissue sections of inoculation sites from NPI-0052-treated and control-treated mice.
  • FIG. 10F compares the effect of NPI-0052 and Bortezomib on tumor growth when administered i.v. to mice.
  • FIG. 10G compares the effect of NPI-0052 and Bortezomib on survival when administered i.v. to mice.
  • FIG. 11A illustrates the effect of NPI-0052 on mitochondrial membrane potential in MM.1S cells incubated with CMXRos.
  • FIG. 11B illustrates the effect of NPI-0052 on superoxide generation in MM.1S cells stained with membrane permeable dye dihydroethidium (HE).
  • FIG. 11C depicts immunoblots of mitochondrial and cytosolic protein fractions obtained from MM.1S cells treated with NPI-0052.
  • FIG. 11D depicts immunoblots of cytosolic proteins obtained from MM.1S cells treated with NPI-0052 and analyzed with anti-caspase-9 Abs.
  • FIG. 11E depicts immunoblots of cytosolic proteins obtained from MM.1S cells treated with NPI-0052 and analyzed with anti-caspase-8 Abs.
  • FIG. 11F depicts immunoblots of MM.1S or MM.1R MM cells treated with NPI-0052 and assessed for apoptosis by both PARP and caspase-3 cleavage assays.
  • FIG. 12A illustrates MM.1S cell viability after treatment with NPI-0052 or Bortezomib in the presence or absence of caspase-3, caspase-8, or caspase-9 inhibitor.
  • FIG. 12B illustrates MM.1S cell viability for cells transfected with vector alone, DN-caspase-8, and DN-caspase-9 after treatment with NPI-0052 or Bortezomib.
  • FIG. 12C depicts immunoblots of cytosolic extracts from DN-caspase-8 and DN-caspase-9 transfected MM.1S cells treated with dexamethasone or anti-Fas MoAb.
  • FIG. 12D illustrates MM.1S cell viability for vector or DN-FADD transfected cells after treatment with NPI-0052 or Bortezomib.
  • FIG. 12E depicts immunoblots of mitochondrial protein extracts from MM.1S MM cells treated with indicated concentration of either NPI-0052 or Bortezomib and analysed with anti-Bax or anti-Hsp60 Abs.
  • FIG. 12F illustrates cell viability of mouse embryonic fibroblasts (MEFs) cells with either wild-type or deleted Bax (knock-out) treated with indicated concentrations of NPI-0052 or Bortezomib.
  • FIG. 13 illustrates viability of normal lymphocytes from five healthy donors treated with indicated concentrations of NPI-0052 or Bortezomib.
  • FIG. 14A illustrates MM.1S cell viability for cells transfected with vector alone or Bcl-2 after treatment with NPI-0052 or Bortezomib.
  • FIG. 14B depicts immunoblots of cytosolic extracts from vector- or Bcl-2-transfected MM.1S cells treated with NPI-0052 or Bortezomib.
  • FIG. 15 illustrates cell viability of MM.1S and MM.1R MM cells treated with indicated concentration of NPI-0052, Bortezomib, or NPI-0052+Bortezomib.
  • prodrugs, metabolites, stereoisomers, and pharmaceutically acceptable salts of the compounds disclosed herein are provided for use as described herein.
  • a “prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • An example, without limitation, of a prodrug would be a compound which is administered as an ester (the “prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • a further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • a prodrug derivative Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs , (ed. H. Bundgaard, Elsevier, 1985), which is hereby incorporated herein by reference in its entirety.
  • pro-drug ester refers to derivatives of the compounds disclosed herein formed by the addition of any of several ester-forming groups that are hydrolyzed under physiological conditions.
  • pro-drug ester groups include pivoyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, as well as other such groups known in the art, including a (5-R-2-oxo-1,3-dioxolen-4-yl)methyl group.
  • Other examples of pro-drug ester groups can be found in, for example, T. Higuchi and V. Stella, in “Pro-drugs as Novel Delivery Systems”, Vol. 14, A.C.S.
  • Metabolites of the compounds disclosed herein include active species that are produced upon introduction of the compounds into the biological milieu.
  • the compounds disclosed herein may exist as a racemate or as enantiomers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present invention.
  • some of the crystalline forms for the compounds of disclosed herein may exist as polymorphs. Such polymorphs are included in one embodiment of the present invention.
  • some of the compounds of the present invention may form solvates with water (i.e., hydrates) or common organic solvents. Such solvates are included in one embodiment of the present invention.
  • pharmaceutically acceptable salt refers to a salt of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • the salt is an acid addition salt of the compound.
  • Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), sulfuric acid, nitric acid, phosphoric acid and the like.
  • compositions can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluensulfonic, salicylic or naphthalenesulfonic acid.
  • organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluensulfonic, salicylic or naphthalenesulfonic acid.
  • Pharmaceutical salts can also be obtained by reacting a compound with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C 1 -C 7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine, and salts with amino acids such as arginine, lysine, and the like.
  • a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C 1 -C 7 alkylamine, cycl
  • compositions which are non-basic, that is, either acidic or neutral excipients.
  • the compounds disclosed herein can be used alone, in combination with other compounds disclosed herein, or in combination with one or more other agents active in the therapeutic areas described herein.
  • halogen atom means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred.
  • esters refers to a chemical moiety with formula —(R) n —COOR′, where R and R′ are independently selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), and where n is 0 or 1.
  • An “amide” is a chemical moiety with formula —(R) n —C(O)NHR′ or —(R) n —NHC(O)R′, where R and R′ are independently selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), and where n is 0 or 1.
  • An amide may be an amino acid or a peptide molecule attached to a molecule of the present invention, thereby forming a prodrug.
  • Any amine, hydroxy, or carboxyl side chain on the compounds of the present invention can be esterified or amidified.
  • the procedures and specific groups to be used to achieve this end are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein in its entirety.
  • purified refers to compounds disclosed herein being free of other, dissimilar compounds with which the compounds of the invention are normally associated in their natural state, so that the compounds of the invention comprise at least 0.5%, 1%, 5%, 10%, or 20%, and most preferably at least 50% or 75% of the mass, by weight, of a given sample.
  • the compound of formula (I) inhibits chymotrypsin-like, trypsin-like, and caspase-like proteasome activities.
  • Bortezomib has been shown to inhibit only chymotrypsin-like proteasome activity. See Goldberg, A. L. & Rock, K. (2002) Nat Med 8, 338-40 and Adams, J. (2004) Nat Rev Cancer 4, 349-60; both of which are incorporated herein by reference in their entirety. It is further demonstrated that compounds of formula (I) have a different mechanism of action than bortezomib.
  • the compound of formula (I) induces apoptosis in various multiple myeloma cell lines including, but not limited to, Dexamethasone-sensitive MM.1S, Dexamethasone-resistant MM.1R, RPMI-8226, OPM2, U266, and Doxorubicin-resistant Dox-40.
  • the compound of formula I also induced apoptosis in cell lines obtained from human multiple myloma patients that had relapsed after multiple prior therapies with Dexamethasone, Bortezomib, and thalidomide.
  • the compound of formula (I) is effective against MM cells that are resistant to other chemotherapeutic agents, including Dexamethasone, Doxorubicin, Bortezomib/PS-341, and thalidomide.
  • a method of treating a neoplastic disease that is susceptible to resistance to at least one chemotherapeutic agent comprising administering to a patient, such as a human, a compound of formula (I) or a pharmaceutically acceptable salt or prodrug ester thereof.
  • a patient such as a human, a compound of formula (I) or a pharmaceutically acceptable salt or prodrug ester thereof.
  • resistance to at least one chemotherapeutic agent it is meant that administration of the chemotherapeutic agent to the patient does not result in significant amelioration of symptoms of the neoplastic disease.
  • “resistance to at least one chemotherapeutic agent” means that administration of the chemotherapeutic agent does not result in appreciable inhibition of the growth of the tumor or reduction in the size of the tumor. “Resistance to at least one chemotherapeutic agent” can also mean that when the agent is exposed to resistant tumor cells, no appreciable apoptosis is induced. By “susceptible to” resistance to at least one chemotherapeutic agent, it is meant that the neoplastic disease currently is resistant to the at least one chemotherapeutic agent or will develop resistance upon repeated administration of the chemotherapeutic agent.
  • a compound of formula (I) when combined with bortezomib trigger synergistic apoptosis in MM cells.
  • a compound of formula (I) may be administered in combination with Bortezomib/PS-341 to achieve apoptosis using lower doses of each agent than if the agents were administered separately, thus reducing the toxicity of the agents.
  • these results demonstrate that a synergistic result may be obtained by administering two different proteasome inhibitors.
  • a method for treating a neoplastic disease comprising administering two or more proteasome inhibitors in synergistic combination.
  • classes of proteasome inhibitors that may be combined include peptide boronate proteasome inhibitors, peptide aldehyde proteasome inhibitors, and non-peptide proteasome inhibitors.
  • a non-limiting example of a peptide boronate proteasome inhibitor is bortezomib.
  • a non-limiting example of a peptide aldehyde proteasome inhibitor is MG-132.
  • Non-limiting examples of non-peptide proteasome inhibitors include omuralide and the compound of formula (I).
  • At least one of the proteasome inhibitors is a compound of formula (I) or bortezomib.
  • the agents are administered simultaneously.
  • administration in combination is accomplished by combining the agents in a single dosage form.
  • the agents are administered sequentially.
  • the agents are administered through the same route, such as orally.
  • the agents are administered through different routes, such as one being administered orally and another being administered i.v.
  • the pharmacokinetics of the two or more agents are substantially the same.
  • a method for treating a neoplastic disease comprising administering a compound of formula (I) in combination with another chemotherapeutic agent.
  • the other chemotherapeutic agent is dexamethasone, doxorubicin, or thalidomide.
  • the other chemotherapeutic agent is another proteasome inhibitor such as bortezomib.
  • a pharmaceutical composition is provided that combines a compound of formula (I) with the additional chemotherapeutic agent.
  • the neoplastic disease treated by any of the methods above may be a cancer selected from breast cancer, sarcoma, leukemia, ovarian cancer, uretal cancer, bladder cancer, prostate cancer, colon cancer, rectal cancer, stomach cancer, lung cancer, lymphoma, multiple myeloma, pancreatic cancer, liver cancer, kidney cancer, endocrine cancer, skin cancer, melanoma, angioma, and brain or central nervous system (CNS) cancer.
  • the neoplastic disease is a multiple myeloma.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising physiologically acceptable surface active agents, carriers, diluents, excipients, smoothing agents, suspension agents, film forming substances, and coating assistants, or a combination thereof, and a compound or combination disclosed herein.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, Pa. (1990), which is incorporated herein by reference in its entirety.
  • Preservatives, stabilizers, dyes, sweeteners, fragrances, flavoring agents, and the like may be provided in the pharmaceutical composition.
  • sodium benzoate, ascorbic acid and esters of p-hydroxybenzoic acid may be added as preservatives.
  • antioxidants and suspending agents may be used.
  • alcohols, esters, sulfated aliphatic alcohols, and the like may be used as surface active agents; sucrose, glucose, lactose, starch, crystallized cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium methasilicate aluminate, synthetic aluminum silicate, calcium carbonate, sodium acid carbonate, calcium hydrogen phosphate, calcium carboxymethyl cellulose, and the like may be used as excipients; magnesium stearate, talc, hardened oil and the like may be used as smoothing agents; coconut oil, olive oil, sesame oil, peanut oil, soya may be used as suspension agents or lubricants; cellulose acetate phthalate as a derivative of a carbohydrate such as cellulose or sugar, or methylacetate-meth
  • composition refers to a mixture of a compound or combination of compounds disclosed herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, injection, aerosol, parenteral, and topical administration.
  • Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • carrier defines a chemical compound that facilitates the incorporation of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • carrier facilitates the uptake of many organic compounds into the cells or tissues of an organism.
  • diot defines chemical compounds diluted in water that will dissolve the compound of interest as well as stabilize the biologically active form of the compound. Salts dissolved in buffered solutions are utilized as diluents in the art.
  • One commonly used buffered solution is phosphate buffered saline because it mimics the salt conditions of human blood. Since buffer salts can control the pH of a solution at low concentrations, a buffered diluent rarely modifies the biological activity of a compound.
  • physiologically acceptable defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
  • compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with suitable carriers or excipient(s).
  • suitable carriers or excipient(s) suitable carriers or excipient(s).
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, topical, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections.
  • parenteral delivery including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections.
  • the compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for prolonged and/or timed, pulsed administration at a predetermined rate.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tabletting processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences, above.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • Suitable excipients are, for example, water, saline, dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like.
  • the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like.
  • Physiologically compatible buffers include, but are not limited to, Hanks's solution, Ringer's solution, or physiological saline buffer. If desired, absorption enhancing preparations (for example, liposomes), may be utilized.
  • penetrants appropriate to the barrier to be permeated may be used in the formulation.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or other organic oils such as soybean, grapefruit or almond oils, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or
  • compositions well known in the pharmaceutical art for uses that include intraocular, intranasal, and intraauricular delivery. Suitable penetrants for these uses are generally known in the art.
  • Pharmaceutical compositions for intraocular delivery include aqueous ophthalmic solutions of the active compounds in water-soluble form, such as eyedrops, or in gellan gum (Shedden et al., Clin.
  • compositions for intranasal delivery may also include drops and sprays often prepared to simulate in many respects nasal secretions to ensure maintenance of normal ciliary action. As disclosed in Remington's Pharmaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, Pa.
  • suitable formulations are most often and preferably isotonic, slightly buffered to maintain a pH of 5.5 to 6.5, and most often and preferably include antimicrobial preservatives and appropriate drug stabilizers.
  • Pharmaceutical formulations for intraauricular delivery include suspensions and ointments for topical application in the ear. Common solvents for such aural formulations include glycerin and water.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a suitable pharmaceutical carrier may be a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • a common cosolvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • VPD co-solvent system is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of POLYSORBATE 80TM; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • hydrophobic pharmaceutical compounds may be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • Agents intended to be administered intracellularly may be administered using techniques well known to those of ordinary skill in the art.
  • such agents may be encapsulated into liposomes. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior.
  • the liposomal contents are both protected from the external micro-environment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm.
  • the liposome may be coated with a tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the desired organ.
  • small hydrophobic organic molecules may be directly administered intracellularly.
  • compositions may be combined with other compositions that contain other therapeutic or diagnostic agents.
  • the compounds or pharmaceutical compositions may be administered to the patient by any suitable means.
  • methods of administration include, among others, (a) administration though oral pathways, which administration includes administration in capsule, tablet, granule, spray, syrup, or other such forms; (b) administration through non-oral pathways such as rectal, vaginal, intraurethral, intraocular, intranasal, or intraauricular, which administration includes administration as an aqueous suspension, an oily preparation or the like or as a drip, spray, suppository, salve, ointment or the like; (c) administration via injection, subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally, intraorbitally, intracapsularly, intraspinally, intrasternally, or the like, including infusion pump delivery; (d) administration locally such as by injection directly in the renal or cardiac area, e.g., by depot implantation; as well as (e) administration topically; as deemed appropriate by those of skill in the art for bringing the
  • compositions suitable for administration include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose.
  • the therapeutically effective amount of the compounds disclosed herein required as a dose will depend on the route of administration, the type of animal, including human, being treated, and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight and mammalian species treated, the particular compounds employed, and the specific use for which these compounds are employed.
  • the determination of effective dosage levels can be accomplished by one skilled in the art using routine pharmacological methods. Typically, human clinical applications of products are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved. Alternatively, acceptable in vitro studies can be used to establish useful doses and routes of administration of the compositions identified by the present methods using established pharmacological methods.
  • dosages may range broadly, depending upon the desired affects and the therapeutic indication. Typically, dosages may be between about 10 microgram/kg and 100 mg/kg body weight, preferably between about 100 microgram/kg and 10 mg/kg body weight. Alternatively dosages may be based and calculated upon the surface area of the patient, as understood by those of skill in the art.
  • compositions of the present invention can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et al. 1975, in “The Pharmacological Basis of Therapeutics”, which is hereby incorporated herein by reference in its entirety, with particular reference to Ch. 1, p. 1).
  • dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient's body weight.
  • the dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient.
  • the present invention will use those same dosages, or dosages that are between about 0.1% and 500%, more preferably between about 25% and 250% of the established human dosage.
  • a suitable human dosage can be inferred from ED 50 or ID 50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.
  • the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity or organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
  • the daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg and 2000 mg of each active ingredient, preferably between 1 mg and 500 mg, e.g. 5 to 200 mg.
  • an intravenous, subcutaneous, or intramuscular dose of each active ingredient of between 0.01 mg and 100 mg, preferably between 0.1 mg and 60 mg, e.g. 1 to 40 mg is used.
  • dosages may be calculated as the free base.
  • the composition is administered 1 to 4 times per day.
  • compositions of the invention may be administered by continuous intravenous infusion, preferably at a dose of each active ingredient up to 1000 mg per day.
  • each active ingredient up to 1000 mg per day.
  • the compounds disclosed herein in certain situations it may be necessary to administer the compounds disclosed herein in amounts that exceed, or even far exceed, the above-stated, preferred dosage range in order to effectively and aggressively treat particularly aggressive diseases or infections.
  • the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compositions should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties may be established by determining in vitro toxicity towards a cell line, such as a mammalian, and preferably human, cell line. The results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • a cell line such as a mammalian, and preferably human, cell line.
  • the results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • the toxicity of particular compounds in an animal model such as mice, rats, rabbits, or monkeys, may be determined using known methods.
  • the efficacy of a particular compound may be established using several recognized methods, such as in vitro methods, animal models, or human clinical trials.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • Dex-sensitive MM.11S and Dex-resistant MM.1R human MM cell lines were obtained from Dr. Steven Rosen (Northwestern University, Chicago, Ill.). See Moalli, P. A., Pillay, S., Weiner, D., Leikin, R. & Rosen, S. T. (1992) Blood 79, 213-22 and Chauhan, D., Catley, L., Hideshima, T., Li, G., Leblanc, R., Gupta, D., Sattler, M., Richardson, P., Schlossman, R. L., Podar, K., Weller, E., Munshi, N. & Anderson, K. C.
  • RPMI-8226 and Doxorubicin (Dox)-resistant (Dox-40) cells were obtained from Dr. William Dalton (Moffit Cancer Center, Tampa, Fla.).
  • U266 and OPM2 MM cell lines were obtained from the American Type Culture Collection (Rockville, Md.).
  • the human tumor cell lines DU 145, HT-29, Jurkat, LoVo, MDA-MB-231, MIA PaCa-2, NCI-H292, OVCAR-3, PANC-1, and PC-3 were purchased from ATCC (Manassas, Va.).
  • MM Cell lines were grown in RPMI-1640 media supplemented with 10% heat inactivated fetal-bovine serum (FBS), 100 units/ml penicillin, 100 ⁇ g/ml streptomycin, and 2 mM L-glutamine. MM cells were freshly isolated from patients relapsing after multiple prior therapies including Dexamethasone (Dex), melphalan, thalidomide or Bortezomib. MM cells were purified from patient bone marrow samples by CD138 positive selection method using CD138 (Syndecan-1) Micro Beads and the Auto MACS magnetic cell sorter (Miltenyi Biotec Inc., Auburn, Calif.).
  • Cell viability and apoptosis assays were assessed by 3-(4,5-dimethylthiozol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Chemicon International Inc., Temecula, Calif.) assay, according to manufacturer's instructions (Roche Molecular Biochemicals, Indianapolis, Ind.), and as described in Chauhan, D., Catley, L., Hideshima, T., Li, G., Leblanc, R., Gupta, D., Sattler, M., Richardson, P., Schlossman, R. L., Podar, K., Weller, E., Munshi, N. & Anderson, K. C. (2002) Blood 100, 2187-94; which is incorporated herein by reference in its entirety. Cell Death Detection ELISAplus was utilized to quantitate cell death, as per manufacturer's instructions (Roche Applied Sciences, Indianapolis, Ind.).
  • the chymotrypsin-like activity of the 20S proteasome was measured as described in Stein, R. L., Melandri, F. & Dick, L. (1996) Biochemistry 35, 3899-908 and Lightcap, E. S., McCormack, T. A., Pien, C. S., Chau, V., Adams, J. & Elliott, P. J. (2000) Clin Chem 46, 673-83; both of which are incorporated herein by reference in their entirety.
  • Purified human erythrocyte-derived 20S proteasome were obtained from Biomol, Plymouth Meeting, Pa.
  • the chymotrypsin-like, caspase-like and trypsin-like activity activities of the 20S proteasome were determined using Suc-LLVY-AMC, Z-LLE-AMC (Boston Biochem, Cambridge, Mass.) and Boc-LRR-AMC (Bachem Bioscience, King of Prussia, Pa.) as peptide substrates, respectively. Fluorescence of the cleaved peptide substrate was measured using a Fluoroskan Ascent 96-well microplate reader (Thermo Electron, Waltham, Mass.). The EC 50 values were calculated by Prism (GraphPad Software) using a sigmoidal dose-response, variable slope model.
  • the EC 50 values were defined as the drug concentration at which 50% of the maximal relative fluorescence is inhibited.
  • the 20S proteasome activity of the individual mice is shown in FIG. 2 with the horizontal bar representing the average activity.
  • Baseline represents the 20S proteasome activity observed in WBC lysates prepared from untreated mice.
  • these findings establish that the compound is orally active and inhibits proteasome activity in vivo.
  • the beta-5 ( ⁇ -5) subunit of the proteasome is markedly less labeled by AdaY( 125 I)Ahx 3 L 3 VS in treated cells than control cells.
  • AdaY( 125 I)Ahx 3 L 3 VS the beta-5 subunit of the proteasome is markedly less labeled by AdaY( 125 I)Ahx 3 L 3 VS in treated cells than control cells.
  • the ⁇ -5 subunit mediates the chymotrypsin-like activity
  • treatment of MM.1S cells with the compound (7 nM) for 6 h also decreased the labeling of the ⁇ -2 subunits (tryptic-activity) and the ⁇ -1 subunits (caspase-like activity) (data not shown).
  • MM cells predominantly localize in the bone marrow microenvironment and their interaction with BMSCs induces production of cytokines which mediate growth of MM cells, as well as protect against drug-induced apoptosis.
  • cytokines which mediate growth of MM cells
  • Positive control shown is an internal control for the assay.
  • Adhesion of MM cells to BMSCs induces IL-6 and IGF-I secretion from BMSCs, which not only regulates the growth of MM cells, but also protects against chemotherapy.
  • BMSCs Adhesion of MM cells to BMSCs induces IL-6 and IGF-I secretion from BMSCs, which not only regulates the growth of MM cells, but also protects against chemotherapy.
  • Reports that high serum levels of IL-6 contribute to clinical chemoresistance and treatment failure, coupled with the ability of the compound of formula (II) (X Cl) to induce MM cell apoptosis even in the presence of IL-6 or IGF-I, suggest that the compound may overcome drug resistance in patients with advanced MM. See Kyrstsonis, M. C., Dedoussis, G., Baxevanis, C., Stamatelou, M. & Maniatis, A. (1996) Br J Haematol 92, 420-422; which is incorporated herein by reference in its entirety.
  • VEGF is elevated in the bone marrow microenvironment and triggers migration, growth, and angiogenesis in MM cells.
  • VEGF induced migration was examined in the presence or absence of the compound (7 or 10 nM).
  • MM.1S cells were stably transfected with Bcl2 construct and analyzed for alterations in cell viability using an MTT assay.
  • Drug treatment was started after the development of measurable tumor. The drug (0.25 mg/kg or 0.5 mg/kg) was given orally twice a week. Serial caliper measurements of perpendicular diameters were done every other day to calculate tumor volume, using the following formula: 4/24 ⁇ (shortest diameter)2 ⁇ (longest diameter). Animals were sacrificed if the tumor was ⁇ 2 cm or necrotic. For tumor growth studies, 7 mice were used in each group.
  • the left panels in FIG. 10B are enlargements of subcutaneous plasmacytomas growing on the right flanks of the mice. Survival was evaluated from the first day of treatment until death; mice were sacrificed when their tumor diameters reached 2 cm or they became moribund ( FIG.
  • mice 10C 10C .
  • concentrations of the compound administered were well tolerated by mice, without evidence of weight loss.
  • Mice in both untreated and treated group were weighed every week. The average changes in the mice body weight are shown in FIG. 10D .
  • Mitochondria play a critical role in apoptosis induction during stress. See Bossy-Wetzel, E. & Green, D. R. (1999) Mutat Res 434, 243-51 and Chauhan, D. & Anderson, K. C. (2003) Apoptosis 8, 337-43; both of which are incorporated herein by reference in their entirety.
  • Superoxide (O 2 ⁇ ) production was measured by staining cells with membrane permeable dye dihydroethidium (HE) for the last 15 min. Superoxide anions oxidize HE to fluorescent ethidium, permitting analysis by flow cytometry.
  • the compound also activates caspase-8 ( FIG. 11E ).
  • caspase-9 mitochondrial-dependent
  • caspase-8 mitochondrial-independent
  • caspase-9 mitochondrial-dependent
  • caspase-8 mitochondrial-independent
  • Total protein lysates were subjected to SDS-PAGE analysis.
  • Immunoblot analysis was performed using antibodies to cytochrome-c, Smac, Caspase-8, -9, or -3 (Cell Signaling, Beverly, Mass.), tubulin (Sigma, St. Louis, Mo.), PARP, Hsp60, or Bax (BD Bioscience Pharmingen, San Diego, Calif.). Blots were developed by enhanced chemiluminesence (ECL; Amersham, Arlington Heights, Ill.).
  • DN dominant-negative
  • Blockade of FADD with DN-FADD significantly attenuated compound of formula (II) (X Cl)-induced cytotoxicity compared to the empty vector-transfected MM.1S cells (42% ⁇ 2.0% viable cells in vector-transfected cells versus 76% ⁇ 5.1% viable cells in DN-FADD-transfected cells; p ⁇ 0.05) ( FIG. 12D ).
  • DN-FADD decreased compound of formula (II) (X Cl)-induced caspase-8 activation; however, minimal caspase-8 activation was still noted (data not shown), which may be due to upstream activators of caspase-8 other than FADD.
  • Bcl-2 During apoptosis Bax neutralizes the antiapoptotic function of Bcl-2, thereby facilitating the cyto-c release and caspase-9 activation.
  • Bcl-2 also confers drug resistance in cancer cells, including MM, and provides partial protection against Bortezomib-induced killing. Therefore, whether ectopic expression of Bcl-2 in MM.1S cells affects the ability of the compound of formula (II) or Bortezomib to trigger cytotoxicity and postmitochondrial apoptotic signaling in MM cells was evaluated.
  • this equation represents the conservation isobologram and indicates additive effects.
  • CI values of ⁇ 1.0 indicate synergism.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US11/293,354 2004-12-03 2005-12-02 Compositions and methods for treating neoplastic diseases Abandoned US20070225350A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US11/293,354 US20070225350A1 (en) 2004-12-03 2005-12-02 Compositions and methods for treating neoplastic diseases
US12/183,007 US8722724B2 (en) 2004-12-03 2008-07-30 Compositions and methods for treating neoplastic diseases
US14/224,965 US10610517B2 (en) 2004-12-03 2014-03-25 Compositions and methods for treating neoplastic diseases
US16/800,570 US20200206190A1 (en) 2004-12-03 2020-02-25 Compositions and methods for treating neoplastic diseases
US17/890,563 US20230210817A1 (en) 2004-12-03 2022-08-18 Compositions and methods for treating neoplastic diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63316104P 2004-12-03 2004-12-03
US11/293,354 US20070225350A1 (en) 2004-12-03 2005-12-02 Compositions and methods for treating neoplastic diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/183,007 Continuation US8722724B2 (en) 2004-12-03 2008-07-30 Compositions and methods for treating neoplastic diseases

Publications (1)

Publication Number Publication Date
US20070225350A1 true US20070225350A1 (en) 2007-09-27

Family

ID=36118298

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/293,354 Abandoned US20070225350A1 (en) 2004-12-03 2005-12-02 Compositions and methods for treating neoplastic diseases
US12/183,007 Active 2028-04-02 US8722724B2 (en) 2004-12-03 2008-07-30 Compositions and methods for treating neoplastic diseases
US14/224,965 Active US10610517B2 (en) 2004-12-03 2014-03-25 Compositions and methods for treating neoplastic diseases
US16/800,570 Abandoned US20200206190A1 (en) 2004-12-03 2020-02-25 Compositions and methods for treating neoplastic diseases
US17/890,563 Pending US20230210817A1 (en) 2004-12-03 2022-08-18 Compositions and methods for treating neoplastic diseases

Family Applications After (4)

Application Number Title Priority Date Filing Date
US12/183,007 Active 2028-04-02 US8722724B2 (en) 2004-12-03 2008-07-30 Compositions and methods for treating neoplastic diseases
US14/224,965 Active US10610517B2 (en) 2004-12-03 2014-03-25 Compositions and methods for treating neoplastic diseases
US16/800,570 Abandoned US20200206190A1 (en) 2004-12-03 2020-02-25 Compositions and methods for treating neoplastic diseases
US17/890,563 Pending US20230210817A1 (en) 2004-12-03 2022-08-18 Compositions and methods for treating neoplastic diseases

Country Status (18)

Country Link
US (5) US20070225350A1 (es)
EP (1) EP1830838B1 (es)
JP (3) JP5676071B2 (es)
KR (1) KR101282191B1 (es)
CN (2) CN101155582B (es)
AU (1) AU2005311709B2 (es)
BR (1) BRPI0517135B1 (es)
CA (1) CA2588923C (es)
DK (1) DK1830838T3 (es)
ES (1) ES2396762T3 (es)
HK (1) HK1113462A1 (es)
IL (3) IL183506A (es)
MX (1) MX2007006526A (es)
NZ (1) NZ555439A (es)
PL (1) PL1830838T3 (es)
SG (1) SG157365A1 (es)
WO (1) WO2006060676A1 (es)
ZA (1) ZA200704402B (es)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070155815A1 (en) * 2002-06-24 2007-07-05 The Regents Of The University Of California Salinosporamides and methods for use thereof
US20080070969A1 (en) * 2004-04-30 2008-03-20 Nereus Pharmaceuticals, Inc. [3.2.0] heterocyclic compounds and methods of using the same
US20080188544A1 (en) * 2007-02-02 2008-08-07 Nereus Pharmaceuticals, Inc. Lyophilized formulations of salinosporamide a
US20090036390A1 (en) * 2004-12-03 2009-02-05 Dana-Farber Cancer Institute Compositions and methods for treating neoplastic diseases
US20090156469A1 (en) * 2007-12-07 2009-06-18 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof in treating waldenstrom's macroglobulinemia
US20090182027A1 (en) * 2003-06-20 2009-07-16 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US20090197937A1 (en) * 2000-11-16 2009-08-06 The Regents Of The University Of California Marine actinomycete taxon for drug and fermentation product discovery
US7579371B2 (en) 2004-04-30 2009-08-25 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US20090298906A1 (en) * 2008-05-12 2009-12-03 Nereus Pharmaceuticals, Inc. Proteasome inhibitors
US20090318529A1 (en) * 2003-06-20 2009-12-24 Fenical William H Salinosporamides and methods for use thereof
WO2010019227A1 (en) * 2008-08-13 2010-02-18 George Mason Intellectual Properties, Inc. Ex vivo therapeutic screening of living bone marrow cells for multiple myeloma
US8003802B2 (en) 2008-03-07 2011-08-23 Nereus Pharmaceuticals, Inc. Total synthesis of Salinosporamide A and analogs thereof
US20130173296A1 (en) * 2011-12-30 2013-07-04 Elwha LLC, a limited liability company of the State of Delaware Evidence-based healthcare information management protocols
US20150299803A1 (en) * 2012-11-05 2015-10-22 Pronai Therapeutics, Inc. Methods of Using Biomarkers for the Treatment of Cancer by Modulation of BCL2 Expression
US10475142B2 (en) 2011-12-30 2019-11-12 Elwha Llc Evidence-based healthcare information management protocols
US10528913B2 (en) 2011-12-30 2020-01-07 Elwha Llc Evidence-based healthcare information management protocols
US10552581B2 (en) 2011-12-30 2020-02-04 Elwha Llc Evidence-based healthcare information management protocols
US10559380B2 (en) 2011-12-30 2020-02-11 Elwha Llc Evidence-based healthcare information management protocols
US10679309B2 (en) 2011-12-30 2020-06-09 Elwha Llc Evidence-based healthcare information management protocols

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6777217B1 (en) 1996-03-26 2004-08-17 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US20030129724A1 (en) 2000-03-03 2003-07-10 Grozinger Christina M. Class II human histone deacetylases, and uses related thereto
US7244853B2 (en) 2001-05-09 2007-07-17 President And Fellows Of Harvard College Dioxanes and uses thereof
AU2005311572A1 (en) * 2004-12-03 2006-06-08 Nereus Pharmaceuticals, Inc. Methods of using (3.2.0) heterocyclic compounds and analogs thereof
WO2006102557A2 (en) * 2005-03-22 2006-09-28 The President And Fellows Of Harvard College Treatment of protein degradation disorders
CA2628110A1 (en) * 2005-11-04 2007-05-18 The Regents Of The University Of California Methods of sensitizing cancer to therapy-induced cytotoxicity
CA2642288C (en) 2006-02-14 2014-10-07 Dana-Farber Cancer Institute, Inc. Histone deacetylase inhibitors
JP5441416B2 (ja) 2006-02-14 2014-03-12 プレジデント アンド フェロウズ オブ ハーバード カレッジ 二官能性ヒストンデアセチラーゼインヒビター
JP2009532484A (ja) 2006-04-06 2009-09-10 ネレアス ファーマシューティカルズ インコーポレイテッド サリノスポラミドa及びその類縁体の全合成
AU2007248656B2 (en) 2006-05-03 2013-04-04 Dana-Farber Cancer Institute, Inc. Histone deacetylase and tubulin deacetylase inhibitors
WO2008124699A1 (en) * 2007-04-06 2008-10-16 Nereus Pharmaceuticals, Inc. A method of using proteasome inhibitors in combination with histone deacetylase inhibitors to treat cancer
US20080280968A1 (en) * 2007-05-04 2008-11-13 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof for treating infectious diseases
WO2009146546A1 (en) * 2008-06-06 2009-12-10 University Health Network 8-hydroxyquinoline derivatives for the treatment of hematological malignancies
US8440716B2 (en) 2008-07-23 2013-05-14 President And Fellows Of Harvard College Deacetylase inhibitors and uses thereof
WO2010099601A1 (en) * 2009-03-05 2010-09-10 University Health Network Use of 5ahq and bortezomib for the treatment of hematological diseases
WO2011019393A2 (en) 2009-08-11 2011-02-17 President And Fellows Of Harvard College Class- and isoform-specific hdac inhibitors and uses thereof
AU2016349833A1 (en) * 2015-11-06 2018-05-24 Accuitis Pharmaceuticals, Inc. Use of proteasome inhibitors to treat ocular disorders
EP3500576A1 (en) 2016-08-19 2019-06-26 Celgene International II Sàrl Morphic forms of marizomib and uses thereof
CA3051846A1 (en) * 2017-01-31 2018-08-09 Carolyn Ruth Bertozzi Agents that inhibit ngly1 and methods of use thereof
CN113750215B (zh) * 2021-09-24 2023-06-23 暨南大学 用于治疗肿瘤的联合药物

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7276530B2 (en) * 2004-04-30 2007-10-02 Nereus Pharmaceuticals, Inc. [3.2.0] Heterocyclic compounds and methods of using the same

Family Cites Families (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5133974A (en) * 1989-05-05 1992-07-28 Kv Pharmaceutical Company Extended release pharmaceutical formulations
US5276182A (en) * 1990-07-09 1994-01-04 The Dow Chemical Company Process for preparing polyurea oligomers
DE4039602A1 (de) * 1990-12-12 1992-06-17 Bauer Kurt Heinz Prof Dr Pharmazeutische formulierungen
KR100221695B1 (ko) * 1991-08-12 1999-09-15 그린 마틴, 브라이언 쥐 테슬리 약학적 구상 제형
US5653962A (en) * 1991-12-12 1997-08-05 Glaxo Group Limited Aerosol formulations containing P134a and particulate medicaments
DK0674510T3 (da) * 1992-11-27 1999-05-10 Napro Biotherapeutics Inc Injicerbart præparat omfattende paclitaxel
US5654286A (en) * 1993-05-12 1997-08-05 Hostetler; Karl Y. Nucleotides for topical treatment of psoriasis, and methods for using same
RU2150942C1 (ru) * 1993-10-01 2000-06-20 Синтекс (Ю.ЭС.ЭЙ.) Инк. Оральные суспензии, содержащие высокие дозы мофетила микофенолята
US5707641A (en) * 1994-10-13 1998-01-13 Pharmaderm Research & Development Ltd. Formulations comprising therapeutically-active proteins or polypeptides
IE80468B1 (en) * 1995-04-04 1998-07-29 Elan Corp Plc Controlled release biodegradable nanoparticles containing insulin
US6838477B2 (en) * 1995-04-12 2005-01-04 President And Fellows Of Harvard College Lactacystin analogs
US6335358B1 (en) * 1995-04-12 2002-01-01 President And Fellows Of Harvard College Lactacystin analogs
US5653987A (en) * 1995-05-16 1997-08-05 Modi; Pankaj Liquid formulations for proteinic pharmaceuticals
US5726181A (en) * 1995-06-05 1998-03-10 Bionumerik Pharmaceuticals, Inc. Formulations and compositions of poorly water soluble camptothecin derivatives
US5667809A (en) * 1995-06-07 1997-09-16 Alliance Pharmaceutical Corp. Continuous fluorochemical microdispersions for the delivery of lipophilic pharmaceutical agents
US5874443A (en) * 1995-10-19 1999-02-23 Trega Biosciences, Inc. Isoquinoline derivatives and isoquinoline combinatorial libraries
US5886210A (en) * 1996-08-22 1999-03-23 Rohm And Haas Company Method for preparing aromatic compounds
IL131410A0 (en) * 1997-02-15 2001-01-28 Proscript Inc Pharmaceutical compositions for treating infarcts through inhibition of nf-kappab
US5922683A (en) * 1997-05-29 1999-07-13 Abbott Laboratories Multicyclic erythromycin derivatives
EP1004573B1 (en) * 1997-08-04 2002-10-30 Taisho Pharmaceutical Co., Ltd Aryloxyaniline derivatives
WO1999009006A1 (en) 1997-08-15 1999-02-25 Soucy Francois SYNTHESIS OF CLASTO-LACTACYSTIN β-LACTONE AND ANALOGS THEREOF
US6133308A (en) * 1997-08-15 2000-10-17 Millennium Pharmaceuticals, Inc. Synthesis of clasto-lactacystin beta-lactone and analogs thereof
US20010051654A1 (en) * 1997-09-25 2001-12-13 Elliott Peter J. Treatment of inflammatory and autoimmune diseases
AU9580098A (en) 1997-09-25 1999-04-12 Millennium Pharmaceuticals, Inc. Proteasome inhibitors, ubiquitin pathway inhibitors or agents that interfere with the activation of nf-(k)b via the ubiquitin proteasome pathway to treat inflammatory and autoimmune diseases
US6831057B2 (en) 1997-10-28 2004-12-14 The University Of North Carolina At Chapel Hill Use of NF-κB inhibition in combination therapy for cancer
DE69842004D1 (de) * 1997-11-21 2010-12-30 Purdue Neuroscience Co Substituierte 2-aminoacetamide und anwendung davon
US6617171B2 (en) 1998-02-27 2003-09-09 The General Hospital Corporation Methods for diagnosing and treating autoimmune disease
JP3794469B2 (ja) * 1998-03-26 2006-07-05 塩野義製薬株式会社 抗ウイルス作用を有するインドール誘導体
US6509331B1 (en) * 1998-06-22 2003-01-21 Elan Pharmaceuticals, Inc. Deoxyamino acid compounds, pharmaceutical compositions comprising same, and methods for inhibiting β-amyloid peptide release and/or its synthesis by use of such compounds
KR20010080267A (ko) 1998-10-20 2001-08-22 밀레니엄 파머슈티컬스 인코퍼레이티드 프로테아솜 억제제 약물 작용을 모니터링하는 방법
FR2784988B1 (fr) * 1998-10-23 2002-09-20 Adir Nouveaux composes dihydro et tetrahydroquinoleiniques, leur procede de preparation et les compositions pharmaceutiques qui les contiennent
US20020049157A1 (en) 1999-08-25 2002-04-25 Jiangping Wu Use of proteasome inhibitors for treating cancer, inflammation, autoimmune disease, graft rejection and septic shock
US20040106539A1 (en) 2000-10-12 2004-06-03 Ulrich Schubert Agents for the treatment of viral infections
US7144723B2 (en) * 2000-11-16 2006-12-05 The Regents Of The University Of California Marine actinomycete taxon for drug and fermentation product discovery
DE10391147D2 (de) * 2002-04-05 2005-02-17 Viromics Gmbh Mittel zur Behandlung von Flaviviridae-Infektionen
US7176232B2 (en) * 2002-06-24 2007-02-13 The Regents Of The University Of California Salinosporamides and methods for use thereof
US7179834B2 (en) * 2002-06-24 2007-02-20 The Regents Of The University Of California Salinosporamides and methods for use thereof
CA2504933C (en) 2002-11-06 2012-10-16 Dana-Farber Cancer Institute, Inc. Methods and compositions for treating cancer using proteasome inhibitors
AU2004212296B2 (en) 2003-02-14 2010-06-17 Intermed Discovery Gmbh Substituted heterocycles
KR20060026052A (ko) * 2003-06-20 2006-03-22 니리어스 파마슈티컬즈, 인코퍼레이션 암, 염증 및 감염성 질환의 치료를 위한 [3.2.0]헤테로사이클릭 화합물 및 그 유사체의 사용방법
ZA200600473B (en) * 2003-06-20 2007-04-25 Univ California Salinosporamides and methods for use thereof
WO2005094423A2 (en) 2004-02-26 2005-10-13 President And Fellows Of Harvard College Selective inhibition of proteasomes of tuberculosis and other bacteria
US7371875B2 (en) * 2004-03-12 2008-05-13 Miikana Therapeutics, Inc. Cytotoxic agents and methods of use
WO2005099687A2 (en) * 2004-04-09 2005-10-27 President And Fellows Of Harvard College Analogs of salinosporamide a
US7183417B2 (en) * 2004-04-09 2007-02-27 President And Fellows Of Harvard College Simple stereocontrolled synthesis of salinosporamide A
US7232818B2 (en) 2004-04-15 2007-06-19 Proteolix, Inc. Compounds for enzyme inhibition
US7579371B2 (en) * 2004-04-30 2009-08-25 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US20060264495A1 (en) * 2004-04-30 2006-11-23 Michael Palladino Methods of using [3.2.0] heterocyclic compounds and analogs thereof
KR20070041742A (ko) 2004-07-12 2007-04-19 바이엘 크롭사이언스 아게 살진균제 및 살충제로서의 치환된 2-피롤리돈 유도체
DK1830838T3 (da) 2004-12-03 2013-01-21 Dana Farber Cancer Inst Inc Sammensætninger og fremgangsmåder til behandling af neoplastiske sygdomme
WO2006060819A2 (en) 2004-12-03 2006-06-08 The Regents Of The University Of California Dhmeq as a sensitizing agent for chemotherapy and immunotherapy of resistant cancer cells
WO2006060609A1 (en) 2004-12-03 2006-06-08 Honeywell International Inc. Gas jet control for inertial measurement unit
AU2005311572A1 (en) * 2004-12-03 2006-06-08 Nereus Pharmaceuticals, Inc. Methods of using (3.2.0) heterocyclic compounds and analogs thereof
WO2006118973A2 (en) 2005-04-29 2006-11-09 Nereus Pharmaceuticals, Inc. Methods of using heterobyclic compounds for treatment of rectal cancer
US20060287520A1 (en) * 2005-05-16 2006-12-21 Danishefsky Samuel J Synthesis of salinosporamide A and analogues thereof
WO2007021897A1 (en) 2005-08-10 2007-02-22 President And Fellows Of Harvard College Analogs of salinosporamide a
WO2007030662A1 (en) * 2005-09-09 2007-03-15 Nereus Pharmaceuticals, Inc. Biosynthesis of salinosporamide a and its analogs
US7465720B2 (en) 2005-09-12 2008-12-16 President And Fellows Of Harvard College Proteasome inhibiting β-lactam compounds
CA2628110A1 (en) * 2005-11-04 2007-05-18 The Regents Of The University Of California Methods of sensitizing cancer to therapy-induced cytotoxicity
GB0605217D0 (en) 2006-03-15 2006-04-26 Novartis Ag Method and compositions for assessing acute rejection
JP2009532484A (ja) 2006-04-06 2009-09-10 ネレアス ファーマシューティカルズ インコーポレイテッド サリノスポラミドa及びその類縁体の全合成
WO2007130404A1 (en) 2006-05-03 2007-11-15 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof for the treatment of lung cancer
DE102006026464A1 (de) 2006-06-01 2007-12-06 Virologik Gmbh Innovationszentrum Medizintechnik Und Pharma Pharmazeutische Zusammensetzung zur Behandlung von Virusinfektionen und / oder Tumorerkrankungen durch Inhibition der Proteinfaltung und des Proteinabbaus
US8088923B2 (en) * 2006-07-07 2012-01-03 The Texas A&M University System Cyclic-fused beta-lactones and their synthesis
US20080280968A1 (en) * 2007-05-04 2008-11-13 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof for treating infectious diseases
US7442830B1 (en) 2007-08-06 2008-10-28 Millenium Pharmaceuticals, Inc. Proteasome inhibitors
US8394816B2 (en) * 2007-12-07 2013-03-12 Irene Ghobrial Methods of using [3.2.0] heterocyclic compounds and analogs thereof in treating Waldenstrom's Macroglobulinemia
CA2717715A1 (en) 2008-03-07 2009-11-05 Nereus Pharmaceuticals, Inc. Total synthesis of salinosporamide a and analogs thereof
JP2011519975A (ja) * 2008-05-12 2011-07-14 ネレアス ファーマシューティカルズ インコーポレイテッド プロテアソーム阻害剤としてのサリノスポラミド誘導体
US8772724B2 (en) 2011-01-05 2014-07-08 Intel-Ge Care Innovations Llc Low power, inexpensive velocity detection using a PIR array

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7276530B2 (en) * 2004-04-30 2007-10-02 Nereus Pharmaceuticals, Inc. [3.2.0] Heterocyclic compounds and methods of using the same

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090197937A1 (en) * 2000-11-16 2009-08-06 The Regents Of The University Of California Marine actinomycete taxon for drug and fermentation product discovery
US8222289B2 (en) 2002-06-24 2012-07-17 The Regents Of The University Of California Salinosporamides and methods for use thereof
US9078881B2 (en) 2002-06-24 2015-07-14 The Regents Of The University Of California Salinosporamides and methods of use thereof
US8637565B2 (en) 2002-06-24 2014-01-28 The Regents Of The University Of California Salinosporamides and methods for use thereof
US10912764B2 (en) 2002-06-24 2021-02-09 The Regents Of The University Of California Salinosporamides and methods of use thereof
US9713607B2 (en) 2002-06-24 2017-07-25 The Regents Of The University Of California Salinosporamides and methods of use thereof
US10314818B2 (en) 2002-06-24 2019-06-11 The Regents Of The University Of California Salinosporamides and methods of use thereof
US7635712B2 (en) * 2002-06-24 2009-12-22 The Regents Of The University Of California Salinosporamides and methods for use thereof
US20070155815A1 (en) * 2002-06-24 2007-07-05 The Regents Of The University Of California Salinosporamides and methods for use thereof
US8217072B2 (en) 2003-06-20 2012-07-10 The Regents Of The University Of California Salinosporamides and methods for use thereof
US20090318529A1 (en) * 2003-06-20 2009-12-24 Fenical William H Salinosporamides and methods for use thereof
US20090182027A1 (en) * 2003-06-20 2009-07-16 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US8168803B2 (en) 2003-06-20 2012-05-01 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US20080070969A1 (en) * 2004-04-30 2008-03-20 Nereus Pharmaceuticals, Inc. [3.2.0] heterocyclic compounds and methods of using the same
US7579371B2 (en) 2004-04-30 2009-08-25 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
US7544814B2 (en) 2004-04-30 2009-06-09 Nereus Pharmaceuticals, Inc. [3.2.0] Heterocyclic compounds and methods of using the same
US8722724B2 (en) 2004-12-03 2014-05-13 Triphase Research And Development I Corp. Compositions and methods for treating neoplastic diseases
US10610517B2 (en) 2004-12-03 2020-04-07 Celgene International Ii Sàrl Compositions and methods for treating neoplastic diseases
US20090036390A1 (en) * 2004-12-03 2009-02-05 Dana-Farber Cancer Institute Compositions and methods for treating neoplastic diseases
US20080188544A1 (en) * 2007-02-02 2008-08-07 Nereus Pharmaceuticals, Inc. Lyophilized formulations of salinosporamide a
US7824698B2 (en) 2007-02-02 2010-11-02 Nereus Pharmaceuticals, Inc. Lyophilized formulations of Salinosporamide A
US20090156469A1 (en) * 2007-12-07 2009-06-18 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof in treating waldenstrom's macroglobulinemia
US8394816B2 (en) 2007-12-07 2013-03-12 Irene Ghobrial Methods of using [3.2.0] heterocyclic compounds and analogs thereof in treating Waldenstrom's Macroglobulinemia
US8003802B2 (en) 2008-03-07 2011-08-23 Nereus Pharmaceuticals, Inc. Total synthesis of Salinosporamide A and analogs thereof
US8314251B2 (en) 2008-03-07 2012-11-20 Nereus Pharmaceuticals, Inc. Total synthesis of salinosporamide A and analogs thereof
US20090298906A1 (en) * 2008-05-12 2009-12-03 Nereus Pharmaceuticals, Inc. Proteasome inhibitors
US8389564B2 (en) 2008-05-12 2013-03-05 Venkat Rami Reddy Macherla Proteasome inhibitors
US7910616B2 (en) 2008-05-12 2011-03-22 Nereus Pharmaceuticals, Inc. Proteasome inhibitors
US20110172285A1 (en) * 2008-05-12 2011-07-14 Nereus Pharmaceuticals, Inc. Proteasome inhibitors
US8227503B2 (en) 2008-05-12 2012-07-24 Nereus Pharmaceuticals, Inc. Proteasome inhibitors
US20110207627A1 (en) * 2008-08-13 2011-08-25 George Mason University Ex vivo therapeutics screening of living bone marrow cells for multiple myeloma
WO2010019227A1 (en) * 2008-08-13 2010-02-18 George Mason Intellectual Properties, Inc. Ex vivo therapeutic screening of living bone marrow cells for multiple myeloma
US10528913B2 (en) 2011-12-30 2020-01-07 Elwha Llc Evidence-based healthcare information management protocols
US10475142B2 (en) 2011-12-30 2019-11-12 Elwha Llc Evidence-based healthcare information management protocols
US10402927B2 (en) 2011-12-30 2019-09-03 Elwha Llc Evidence-based healthcare information management protocols
US10552581B2 (en) 2011-12-30 2020-02-04 Elwha Llc Evidence-based healthcare information management protocols
US10559380B2 (en) 2011-12-30 2020-02-11 Elwha Llc Evidence-based healthcare information management protocols
US20130173296A1 (en) * 2011-12-30 2013-07-04 Elwha LLC, a limited liability company of the State of Delaware Evidence-based healthcare information management protocols
US10679309B2 (en) 2011-12-30 2020-06-09 Elwha Llc Evidence-based healthcare information management protocols
US20150299803A1 (en) * 2012-11-05 2015-10-22 Pronai Therapeutics, Inc. Methods of Using Biomarkers for the Treatment of Cancer by Modulation of BCL2 Expression

Also Published As

Publication number Publication date
JP2015028089A (ja) 2015-02-12
US20090036390A1 (en) 2009-02-05
BRPI0517135B1 (pt) 2022-04-19
EP1830838B1 (en) 2012-10-03
BRPI0517135A (pt) 2008-09-30
JP5676071B2 (ja) 2015-02-25
US8722724B2 (en) 2014-05-13
US20200206190A1 (en) 2020-07-02
MX2007006526A (es) 2007-09-19
KR101282191B1 (ko) 2013-07-08
PL1830838T3 (pl) 2013-07-31
CN101155582B (zh) 2013-04-24
KR20080003306A (ko) 2008-01-07
IL236426B (en) 2019-11-28
NZ555439A (en) 2010-12-24
CN101155582A (zh) 2008-04-02
AU2005311709B2 (en) 2011-07-28
US10610517B2 (en) 2020-04-07
EP1830838A1 (en) 2007-09-12
JP2012236862A (ja) 2012-12-06
SG157365A1 (en) 2009-12-29
ES2396762T3 (es) 2013-02-26
CA2588923C (en) 2015-02-03
WO2006060676A1 (en) 2006-06-08
US20230210817A1 (en) 2023-07-06
JP2008521928A (ja) 2008-06-26
JP5837469B2 (ja) 2015-12-24
CN103230394B (zh) 2016-04-20
IL236426A0 (en) 2015-02-26
HK1113462A1 (en) 2008-10-03
US20140349965A1 (en) 2014-11-27
ZA200704402B (en) 2008-07-30
CN103230394A (zh) 2013-08-07
CA2588923A1 (en) 2006-06-08
IL183506A0 (en) 2007-10-31
JP5963830B2 (ja) 2016-08-03
AU2005311709A1 (en) 2006-06-08
IL241756A0 (en) 2015-11-30
DK1830838T3 (da) 2013-01-21
IL183506A (en) 2015-10-29

Similar Documents

Publication Publication Date Title
US20230210817A1 (en) Compositions and methods for treating neoplastic diseases
Chauhan et al. A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib
EP2796134B1 (en) Combination of the peptide epoxyketone proteasome inhibitor carfilzomib with melphalan for use in the treatment of multiple myeloma
EP2263694A1 (en) Antitumor agent comprising the histone deacetylase inhibitor FK228 and a topoisomerase II inhibitor
KR102358632B1 (ko) 스트렙토니그린 및 항암제를 포함하는 대장암 예방 또는 치료용 약학적 조성물
EP1280527B1 (en) Use of substituted acryloyl distamycin derivatives in the treatment of tumors associated with high levels of glutathione
AU2001260227A1 (en) Use of substituted acryloyl distamycin derivatives in the treatment of tumors associated with high levels of glutathione
CN112972684B (zh) Iap抑制剂与parp或mek抑制剂或其他化学治疗剂的组合
WO2021110011A1 (en) Combination of iap inhibitors and parp or mek inhibitors or other chemotherapeutic agents
EP3390408A1 (en) Novel compounds and methods for the treatment of alzheimer's disease and/or cerebral amyloid angiopathy
MXPA06009577A (es) Agente antitumoral

Legal Events

Date Code Title Description
AS Assignment

Owner name: DANA-FARBER CANCER INSTITUTE, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ANDERSON, KENNETH C.;CHAUHAN, DHARMINDER;REEL/FRAME:017456/0915;SIGNING DATES FROM 20060327 TO 20060328

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:DANA-FARBER CANCER INST;REEL/FRAME:036528/0569

Effective date: 20150902

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH-DIRECTOR DEITR, MARY

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:DANA-FARBER CANCER INSTITUTE;REEL/FRAME:040144/0422

Effective date: 20160926

AS Assignment

Owner name: TRIPHASE RESEARCH AND DEVELOPMENT I CORP., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEREUS PHARMACEUTICALS, INC.;REEL/FRAME:041698/0404

Effective date: 20121023