WO2007092436A2 - Compounds for treating inflammatory disorders, demyelinating disorders and cancers - Google Patents

Compounds for treating inflammatory disorders, demyelinating disorders and cancers Download PDF

Info

Publication number
WO2007092436A2
WO2007092436A2 PCT/US2007/003135 US2007003135W WO2007092436A2 WO 2007092436 A2 WO2007092436 A2 WO 2007092436A2 US 2007003135 W US2007003135 W US 2007003135W WO 2007092436 A2 WO2007092436 A2 WO 2007092436A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
optionally substituted
methyl
taken together
independently
Prior art date
Application number
PCT/US2007/003135
Other languages
French (fr)
Other versions
WO2007092436A3 (en
Inventor
Alfred M. Ajami
Original Assignee
Xanthus Pharmaceuticals, 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 Xanthus Pharmaceuticals, Inc. filed Critical Xanthus Pharmaceuticals, Inc.
Priority to US11/893,375 priority Critical patent/US20080108641A1/en
Publication of WO2007092436A2 publication Critical patent/WO2007092436A2/en
Publication of WO2007092436A3 publication Critical patent/WO2007092436A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/06Peri-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • R a and R b are not H or optionally substituted alkyl and R y is not optionally substituted N-morpholinyl, optionally substituted N-piperazinyl, or optionally substituted N-pyrazinyl.
  • R x when R is R x , then R a and R b are not H or optionally substituted alkyl and R y is not optionally substituted N-morpholinyl, or optionally substituted N- pyrazinyl.
  • FIG. 7 is a time dependent chart showing mean clinical score (performance score) of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with SymadexTM after 8 weeks of dosing at 20 mg/kg versus vehicle control.
  • EAE Experimental Autoimmune Encephalomyelitis
  • FIG.8B is a bar chart showing the time course of CD-4 cell counts of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with SymadexTM after 4, 6 and 8 weeks of dosing at 20 mg/kg versus vehicle control.
  • EAE Experimental Autoimmune Encephalomyelitis
  • FIG.8D is a bar chart showing the time course of B-cell counts of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with SymadexTM after 4, 6 and 8 weeks of dosing at 20 mg/kg versus vehicle control.
  • EAE Experimental Autoimmune Encephalomyelitis
  • FIG. 13 is a time dependent chart bar chart showing mean performance of the animals suffering from acute stage Collagen Monoclonal Antibody (mAB) Induced Arthritis at the indicated day post treatment with SymadexTM after 3 consecutive daily oral doses at 30 mg/kg.
  • mAB Collagen Monoclonal Antibody
  • R y is a heteroaryl or a non-aromatic heterocycle, each optionally substituted at one or more substitutable carbons with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d , wherein R c and R d are individually H, methyl or ethyl.
  • R a and R b are not H or optionally substituted alkyl and R y is not optionally substituted N-morpholinyl, optionally substituted N-piperazinyl, or optionally substituted N-pyrazinyl.
  • R is selected from groups represented by structural formulas (II) - (VII):
  • R 21 is optionally substituted Cl-ClO alkyl, or an optionally substituted aryl or aralkyl or, R 21 and R 22 taken together with their intervening atoms form a 5-7 membered non-aromatic heterocycle.
  • R 22 is not a part of a heterocycle defined above, the R and R are each independently -H, or optionally substituted C1-C6 alkyl, provided that R 22 and R 23 are not simultaneously hydrogens.
  • R 21 is optionally substituted Cl-ClO alkyl, phenyl or benzyl optionally substituted with a halogen, -NO 2 ..
  • R 22 and R 23 are each independently -H, or a C1-C3 alkyl; or R 21 and R 22 , taken together with their intervening atoms, form a 5 or 6 membered non-aromatic heterocycle and R 23 is -H, or a Cl -C3 alkyl.
  • the remainder of the variables take the values defined above in formula (I).
  • R 101 is H, optionally substituted Cl -C6 alkyl or optionally substituted aryl or aralkyl.
  • R 100 is phenyl or benzyl optionally substituted with a halogen, -NO 2 , -NH 2 . -COOH, alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, Cl- C3 haloalkyl or C1-C3 haloalkoxy.
  • R !0! is H or C1-C4 alkyl. More preferably, R 101 is H 5 methyl or ethyl. The remainder of the variables take the values defined above in formula (I).
  • R 107 is C1-C6 alkyl, optionally substituted aryl or aralkyl, or a non -aromatic heterocycle, optionally substituted at one or more substirutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d .
  • R alone or taken together with R 4 , or alternatively R 5 , and the intervening carbon atoms is a phenol isosteric group.
  • phenol isosteric group means a chemical moiety whose quantum properties result in such electrostatic charge distribution, polarizability, capacity to form hydrogen bonds, hydrophobicity, steric effect and other inductive or mesomeric effects, that the molecule as a whole is endowed with activity as either agonist or antagonist of receptors, enzyme active sites, and protein/enzyme allosteric modulation sites in the context of biological function.
  • alkyl as used herein, unless otherwise indicated, includes straight or branched saturated monovalent hydrocarbon radicals, typically Cl-ClO, preferably C1-C6.
  • alkyl groups include, but are not limited to, methyl, " ethyl, propyl, isopropyl, and t-butyl.
  • cycloalkyl is a non-aromatic saturated carbocyclic moieties.
  • examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • Suitable substituents for a cycloalkyl. are defined above for an alkyl.
  • aryl refers to a carbocyclic aromatic group. Examples of aryl groups include, but are not limited to phenyl and naphthyl.
  • heteroaryloxy means a “heteroaryl-O-" group, wherein heteroaryl is defined above.
  • non-aromatic heterocyclic rings examples include 3-tetrahydrofuranyl, 2-tetrahydropyranyl, 3-tetrahydropyranyl, 4-tetrahydropyranyl, [l,3]-dioxalanyl, [l,3]-dithiolanyl, [1,3]- dioxanyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3- morpholinyl, 4-morpholinyl, 2-thiomorphoIinyl, 3-thiomorpholinyl, 4- thiomorpholinyl, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3-pyrorolidinyl, 1 -piperazinyl, 2- piperazinyl, 1 -piperidinyl, 2- ⁇ i ⁇ eridinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, N-substi
  • the "activated alkanoic acylating agent" is defined within the references cited.
  • This acetimidate reacts with the amine in compound (S 1.3) and cyclized to the methyl or ethyl or other alkylimidazole, again per the cited articles.
  • Examples of chronic inflammatory demyelinating neuropathies include: chronic Immune Demyelinating Polyneuropathy (CIDP); multifocal CIDP; multifocal motor neuropathy (MMN); anti-MAG Syndrome (Neuropathy with IgM binding to Myelin-Associated Glycoprotein); GALOP Syndrome (Gait disorder Autoantibody Late-age Onset Polyneuropathy); anti-sulfatide antibody syndrome; anti-GM2 gangliosides antibody syndrome; POEMS syndrome (Polyneuropathy Organomegaly Endocrinopathy or Edema M-protein Skin changes); perineuritis; and IgM anti-GDlb ganglioside antibody syndrome.
  • the method comprises administering to a patient a therapeutically effective amount of a compounds of formula (I) or a pharmaceutically acceptable salt thereof.
  • the present invention is a method of treatment of a patient suffering from a demyelinating condition.
  • a demyelinating condition is a condition that destroys, breaks the integrity of or damages a myelin sheath.
  • myelin sheath refers to an insulating layer ' surrounding vertebrate peripheral neurons, that increases the speed of conduction and formed by Schwann cells in- the peripheral or by oligodendrocytes in the central nervous system.
  • the present invention is a method of reversing paralysis in a subject in need thereof with a demyeliriating disease, comprising administering to the subject a compound in an amount sufficient to inhibit lymphocyte infiltration of immune cells in the spinal cord to promote remyelination of nerve cells in the spinal cord and .thereby treating paralysis in said subject, wherein the compound is of formula formula (I) or a pharmaceutically acceptable salt thereof.
  • the compounds of formula (I) are administered orally in the amount of 1-3 mg/kg daily for 10-15 days, repeating every 30-45 days.
  • the compounds of formula (I) are administered several times over a period of up to 40-60 days, up to a cumulative dose of from 10 mg/kg to 45 mg/kg. Administration can be repeated over one or more periods of up to 40-60 days.
  • the administration of the compounds or the combinations of the compounds described herein results in an effective blood level of the compound in the patient of more than or equal to 10 ng/ml.
  • compounds can be administered intravenously in an amount of 20 ⁇ g to about 500 ⁇ g per kilogram body weight of the patient.
  • the term “combination” with reference to pharmaceutically active agents and the term “co-administering” and “co-administration” refer to administering more than one pharmaceutically active agent to a patient during one- treatment cycle and not necessarily simultaneous or in a mixture.
  • Suitable anti -inflammatory agents include corticosteroid such as prednisone, methylprednisolone, dexamethasone Cortisol, cortisone, fludrocortisone, prednisolone, 6 ⁇ -methylprednisolone, triamcinolone, or betamethasone.
  • the compounds of the present invention can be administered in combination with one or more other pharmaceutically active agents that are effective against multiple sclerosis.
  • agents include the interferons (interferon beta 1-a, beta 1-b, and alpha), g ⁇ atiramer acetate or corticosteroids such as methylprednisolone and prednisone as well as chemotherapeutic agents such as mitoxantrone, methotrexate, azathioprine, cladribine cyclophosphamide, cyclosporine and tysabri.
  • compositions of the present invention include: 3-4 diaminopyridine; ABT-874; Actos® (pioglitazone); ALCAR (acetyl-L- carnitine); Alpha lipoic acid; AndroGel® (testosterone gel); combination of trimethoprim and vitamin C; combination of azithromycin and rifampin; minocycline; donezepil HCL; Avandia® (rosiglitazone maleate; combination of IFN beta-la) and acetaminophen, ibuprofen or prednisone; combination of Avonex® (interferon beta- Ia) + CellCept®
  • pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include compounds include: Mylinax, an oral formulation of cladrlbine used in leukaemia treatment, developed by Serono/Ivex; Teriflunomide.
  • methyl donator developed by Transition Therapeutics
  • Laquinlmod an oral formulation of a derivative of linomide, developed by Active Biotecb/Teva
  • deskar pirfenidone ⁇ a TNF -alpha inhibitor developed by Mamac
  • ATL-1102 a second generation antisense inhibitor targeting VLA4, developed by Antisense Therapeutics. . •
  • compounds of formula (A) can be administered in combination with antivascular agents, in particular agents inhibiting the growth " factor receptors, Epidermal Growth Factor Receptor. (EGFR), Vascular Epidermal Growth Factor Receptor (VEGFR), and Fibroblast Growth Factor Receptor (FGFR).
  • antivascular agents in particular agents inhibiting the growth " factor receptors, Epidermal Growth Factor Receptor. (EGFR), Vascular Epidermal Growth Factor Receptor (VEGFR), and Fibroblast Growth Factor Receptor (FGFR).
  • the present invention is a method of treating a subject suffering from, a cancer.
  • cancer refers to the uncontrolled growth of abnormal cells that have mutated from normal tissues.
  • a cancerous tumor (malignancy) is of potentially unlimited growth and expands locally by invasion and systemically by metastasis.
  • cancers that can be treated by the compounds of the present invention include: colorectal cancer, non- small cell lung cancer, ovarian, renal, sarcoma, melanoma, head & neck, hepatocellular, thyroid, multidrug-resistant leukemia, lymphoma, and multiple myeloma.
  • Treating a subject suffering from cancer includes achieving, partially or substantially, one or more of the following: arresting the growth or spread of a cancer, reducing the extent of a cancer (e.g., reducing size of a tumor or reducing the number of affected sites), inhibiting the' growth rate of a cancer, and ameliorating or improving a clinical symptom or indicator associated with a cancer (such as tissue or serum components).
  • Treating refers to partially or totally inhibiting, delaying, or reducing (partially or completely) the progression of cancer including cancer metastasis; inhibiting, delaying or preventing the recurrence of cancer including cancer metastasis in,. for example, a human.
  • sarcomas cancers arising from connective or supporting tissues such as bone or muscle
  • carcinomas cancers arising from the body's glandular cells and epithelial cells, which line body tissues
  • lymphomas cancers of the lymphoid organs such as the lymph nodes, spleen, and thymus, which produce and store infection-fighting cells.
  • ovarian cancer is cancer of the ovaries or fallopian tubes, including cancers of germ cells, stromal cells, and epithelial cells.
  • ovarian cancers include but are not limited to:
  • Epithelial Ovarian Tumors which include but are not limited to, serous adenomas, mucinous adenomas, and Brenner tumors, rumors of low malignant potential (LMP tumors), borderline epithelial ovarian cancer, epithelial ovarian cancers, carcinomas and undifferentiated epithelial ovarian carcinomas;
  • Germ Cell tumors which include but are not limited to, teratoma, dysgerminoma, endode ⁇ al sinus tumor, and choriocarcinoma; and
  • Renal cancer or “kidney cancer”, as used herein, includes but is not limited to, transitional cell cancer (TCC) of the renal pelvis, Wilms Tumour and renal cell cancer.
  • TCC transitional cell cancer
  • Renal cell cancer is also called renal adenocarcinoma or hypernephroma.
  • the cancerous cells are found in the lining of the tubules (the smallestjubes inside the nephrons that help filter the blood and make urine).
  • renal cell cancer There are several types of renal cell cancer including but not limited to clear cell, chromophilic, chromophobic, oncocytic, collecting duct and sarcomatoid.
  • Renal cancer also includes cancers containing more than one of the cell types described above.
  • melanoma is a type of skin cancer that occurs in the cells that color the skin, called melanocytes.
  • Types of melanoma include but are not limited to: Cutaneous melanoma, superficially spreading melanoma, nodular malignant melanoma, lentiginous malignant melanoma, acral lentiginous melanoma, demoplastic malignant melanomas ' , giant melanocyte nevus, amelandtic malignant melanoma, acral lentiginous melanoma unusual melanoma variants, including mucosal malignant melanoma and ocular malignant 'melanoma.
  • Sphercomas include but are not limited to, fibrosarcomas from fibrous body tissues, leiomyosarcomas and rhabdomyosarcomas from muscle tissues, liposarcomas from fat, synovial sarcomas, angiosarcomas from blood vessels, MPNST - malignant peripheral nerve sheath tumours (PNSTs), GIST - gastrointestinal stromal sarcoma, osteosarcoma, myosarcoma, chondrosarcoma, bile duct sarcoma, brain sarcoma, breast sarcoma, soft tissue sarcoma, uterine sarcoma, endocardial sarcoma, stromal sarcomas from supporting tissues (endometrial stromal sarcoma), granuloytic, histiolytic, hemangioendothelial, Kupffer-cell, neurogenic, round-
  • thyroid cancer includes but is not limited to, papillary and/or mixed papillary/follicular, follicular and/or Hurthle cell, lymphoma, medullary, anaplastic and combinations thereof.
  • cancers include but are not limited to squamous cell carcinoma, mucoepidermoid carcinoma, adenoid cystic carcinoma, lymphoma, adenocarcinoma, esthesioneuroblastoma, tumors of the nasal cavity and paranasal sinuses, nasopharyngeal cancer, cancers of the oral cavity (including all the various parts of the mouth: the lips; the lining inside the lips and cheeks (the buccal mucosa); the bottom of the mouth; the front of the tongue; the front part of the top of the mouth (the hard palate); the gums; and the area behind the wisdom teeth (the retromolar trigone)), tumors of the oropharynx, hypopharyngeal tumors, laryngeal cancer and salivary gland cancer (including malignant salivary gland tumor).
  • non-small, cell lung cancer includes, squamous cell . carcinoma, adenocarcinoma and undifferentiated non-small cell lung cancer (undeveloped cancer cells are known as undifferentiated cells) and large cell carcinoma.
  • the present invention is a method of treating a patient suffering from an acute myeloid leukemia characterized by a FLT3 mutation.
  • the compound of formula (I) can be administered in combination with an anti-cancer agent.
  • the compounds used in the present invention can be administered alone or in combination with one or more other pharmaceutically active agents that are effective against the cancer being treated.
  • anti-cancer drugs that can be employed in combination with the compounds described herein include: 20-epi-l,25 dihydroxyvitamin D3; 5- ethynylurac.il; abiraterone; aclar ⁇ bicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; ainidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; ' andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti- dorsalizing morphogenetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apuri
  • xhemotherapeutic agents that can be employed in combination with the compounds of the invention include but are not limited to alkylating agents, antimetabolites, natural products, or hormones.
  • the compound of formula (I) of this invention may also be administered topically, and when done so the carrier may suitably comprise a solution, ointment or gel base.
  • the base for example, may comprise one or more of petrolatum, • lanolin, polyethylene glycols, bee wax, mineral oil,- diluents such as water and alcohol, and emulsif ⁇ ers and stabilizers.
  • IL-4 serves as a growth and differentiation factor for B cells, mast cells and macrophages and is a switch factor for synthesis of IgE in mice. It also promotes growth of a cloned CD4 + T cell and enhances class II MHC molecule expression and resting B lymphocytes enlargement. In man, CD4 + T lymphocytes also produce IL- 4, but the human variety has not been shown to serve as a B cell or mast cell growth factor. Both murine and human IL-4 induce switching of B lymphocytes to synthesize IgE. Human IL-4 also induces CD23 expression by B lymphocytes and macrophages in man. IL-4 may have some role in cell mediated immunity.
  • IL-IO inhibits cytokine synthesis by T H I cells, blocks antigen presentation, and inhibits the formation of interferon ⁇ .
  • IL-10 inhibits the macophage's ability to present antigen and to form IL-I, IL-6 and TNF- ⁇ .
  • IL-10 also participates in IgE regulation. Although IL-10 suppresses cell-mediated immunity, it stimulates B lymphocytes, IL-2 and IL-4 T lymphocyte responsiveness in vitro, and murine mast cells exposed to IL-3 and IL-4.
  • IL-10 may find therapeutic utility by suppressing T lymphocyte autoimmunity in multiple sclerosis and type I diabetes mellitus as well as in facilitating allograft survival.
  • T-lymphocyte cells isolated from thymus of balb/c mice weighing 17 ⁇ 1 g were used.
  • Test compound and/or vehicle is incubated with the cells (4 x 10 6 /ml) in the presence of 3 ⁇ g/mL Concanavalin A (Con A) in AIM-V medium pH 7.4 at 37 0 C for 24 hours.
  • Concanavalin A Con A
  • [ 3 H]Thyrmdine (120 nM) was then added for an additional overnight incubation period. Thymidine incorporation was assessed by liquid scintillation counting.
  • Treatments of EAE come in many structural forms: treatment can be prophylactic or preventative, whereby the therapeutic composition is administered before immunization; treatment can be initiated during the first week of induction; and treatment can be interventious, initiated after clinical symptoms are extent (acute or chronic).
  • Prevention protocols are very common in the literature, treatment after disease is rarer, and treatment after weeks of disease are the most infrequent.
  • the experiments reported herein are in the last classification in which animals in the chronic- progressive (CP) phase with extensive demyelinated plaques are treated.
  • CP- EAE induced by whole CNS in complete Freund's adjuvant is a florid disease with extensive inflammatory and demyelinated changes.
  • CP- EAE induced by whole CNS in complete Freund's adjuvant is a florid disease with extensive inflammatory and demyelinated changes.
  • As a general philosophy we believe that successful intervention at later times can better predict effectiveness in the human condition. This is particular relevant to the case of prevention studies, which concentrates on the peripheral immune system,
  • H-E hematoxylin-eosin
  • SCR solochrome-R-cyanin
  • FIG. 4 The longitudinal course of recovery from disease is further illustrated in FIG.” 4.
  • the vehicle controls showed a steady course of disease.
  • Treated animals, in both the 20 and 40 mg/kg cohorts entered the study presenting disease of greater severity than controls, a coincidental circumstance attributed to the phenomenology of randomization prior to assignment of a treating group.
  • treatment began on day forty and progressed for a total of four doses.
  • both treated cohorts showed significant disease improvement to levels significantly below control, despite having entered the study at a disease level well above control.
  • Statistical significance even with 3 and 4 animal small cohorts showed p values supporting the hypothesis that treatment afforded a statistically different outcome over control. These were determined by non-parametric comparisons by the Mann- Whitney or Wilcoxon rank-sum test for difference in medians. The significance values were 0.001 and 0.004 for the 40 and 20 mg/kg cohorts, respectively.
  • Demyelination is a key pathological feature of the MS lesion..Not only does this alter electrical response of the axon, current thought suggests that prolonged demyelination can result in permanent axonal damage and death. Thus neuro- degeneration is also a key component of the MS pathological milieu.
  • SymadexTM has proved to be effective in permitting endogenous remyelination even after a period of disease progression that reached 97% spinal chord demyelination in this chronic - progressive model. • It appears to permit this CNS recovery by reducing the inflammation in existing lesions.
  • Example 2 The only significant protocol deviation from the method of Example 2 was that the pool of immunized animals was culled of animals presenting with a ' disease severity greater than 2 and randomized so that the' mean disease severity of each cohort was matched in the severity score range of 1 to 1.5. This measure was invoked in order to avoid the chance circumstance, observed in Example 2, that animal selected for treatment should start treatment with a more severe presentation than the corresponding vehicle controls.
  • AU treated animals showed statistically significant improvement in disease. That is, their symptoms of paralysis attributable to the demyelinating progression of inflammatory cell assault on nerve chord parenchyma, were reduced close to baseline, pre-disease levels. These results are evident by mere inspection of the disease course plots and also proved to be highly significant by non-parametric, rank-order statistical analysis.
  • the therapeutic response shows a graded temporal response against control. Sick animals become progressively healthier in proportion to the weekly duration of response, while . control animals progress irreversibly towards complete neurological dysfunction, whose root cause is irreversible demyelination.
  • the cumulative pharmacodynamic effect of drug treatment is durable, because treated animals remain in stable condition, commensurate with their degree of treatment, while untreated controls sustain an accelerating progression in. disease. This is a particularly noteworthy observation in light of the effects obtained with the ' most promising recent therapy for progressive MS, namely the ⁇ 4 integrin antagonists like Tysabri and its small molecule ligand equivalents, as described by Piraino PiS. et al, J.
  • Symadex does not exert its action via the activation and recruitment of inflammatory cells.
  • the histopathology of • spinal chord from animals sacrificed at periodic interval throughout the time course of disease recovery show accumulation, rather than diminution, of inflammatory cells in blood vessels and perivascular cuffs, as noted in Example 2. Yet, these cells are apparently blocked from transmigrating beyond the basement membrane of parenchyma, suggesting a block via mechanisms that could involve: cell adhesion, motility, and extracellular matrix remodeling.
  • Experiment 2 was applied to a cohort of animals and controls, which were allowed to reach ' the chronic phase of disease at.30 days post immunization. Six animals with a disease score of 1 were selected and half were treated -with 20 mg/kg SymadexTM administered intraperitoneally. Two dose were given 72 hours apart to 3 animals. Three animals served as vehicle controls.
  • the SymadexTM treatment on a 72 hour schedule reversed the progression of disease and restored baseline clinical scores with just 2 doses, while disease progression continued in the control cohort.
  • the difference is statistically significant by a p value of 0.002 by the rank-sum test.
  • Example 5 SymadexTM Alleviates the Symptoms of Experimental Autoimmune Encephalomyelitis (EAE), an Animal Model of Acute Multiple Sclerosis Upon Daily Dosing Over the Initial Course of Disease Induction.
  • EAE Experimental Autoimmune Encephalomyelitis
  • the EAE model in the guinea pig is biphasic.
  • the typical clinical pattern of neurological impairment begins with acute signs of disease day 9 post immunization.
  • Clinical onset results in weight loss, hind limb weakness and an abnormal righting reflex.
  • the severity of these symptoms peaks over 6-7 additional days followed by a short duration transient and partial resolution by day until day- 20, when the disease course changes to a steady progressive decline, from which there is no clinical recovery.
  • Example 4 As an important extension to the utility of Symadex, its therapeutic effect at this earlier stage of disease presentation was examined. The experiment was further designed to build on the results of Example 4, which suggested that more frequent dosing affords more rapid and unidirectional symptom resolution. Since the acute phase of EAE also mimics active, but not necessarily progressive disease, as would be expected to be the case in human subjects with remitting-relapsing multiple sclerosis, the experiment was further designed to test the efficacy in comparison to mitoxantrone. This later drug, as indicated earlier, is an approved therapeutic agent and had served as the starting point for the chemical evolution of what became the SymadexTM molecule minus the toxicophoies known to be causative agents for cardiotoxicity.
  • the mitoxantrone .dose was selected to reflect a typical high dose given to rats or mice by daily dosing in the prior art, but allonietrically scaled to the guinea pig-
  • the cohorts treated with either SymadexTM or mitoxantrone present a consistently different trend than the vehicle controls.
  • the onset of disease is followed by a steady increase in clinical score until day 15, followed by the characteristic short term reversion and a second climb towards higher disease severity by day 20.
  • mitoxantrone and Symadex the initial rise in neurological impairment is arrested, and all animals continued on a course of recovery towards basal levels throughout the dosing period.
  • FIG. 11 shows the weight gain profile, a sensitive indicator of general health status in guinea pigs. Acute EAE disease onset triggers a rapid weight loss from which there develops a steady recovery after day-15. Control animals and SymadexTM treated animals regain the ability to add weight every day, which is the norm for guinea pigs when healthy and prior to the onset of severe, chronic disease.
  • mitoxantrone may have alleviated the acute clinical symptoms of EAE at a lower relative dose but it also impairs weight gain, a sign of generalized toxicological response to a drug that is a potent and broad spectrum cytotoxic.
  • Comparison of the weight gain profiles between vehicle controls and SymadexTM did not show statistical significance by the Mann- Whitney rank-sum test, whereas the difference between SymadexTM and mitoxantrone reached statistical significance with a p value of 0.034. .
  • RA Rheumatoid Arthritis
  • DMARDS disease modifying antirheumatic drugs
  • TNF- ⁇ tumor necrosis factor ⁇
  • MTX methotrexate
  • CIA Collagen-induced Arthritis
  • rodents rat and mouse
  • nonhuman primates by immunization with type II collagen, the major constituent protein of articular cartilage.
  • CIA manifests as swelling and erythema in the limbs of the mouse.
  • This model of autoimmunity shares several clinical and pathological features with rheumatoid arthritis (RA) and has become the most widely studied model of RA.
  • CIA in the mouse model was first described by Courtenay et al. in 1980 (Courtnay, J.S., Dallman, MJ., Dayman, A.D., Martin A. s and Mosedale, B.
  • mice Groups of 3 BALB/c strain mice, 6-7 weeks of age, were used for the induction of arthritis by monoclonal antibodies (mABs) raised against type II collagen, plus Iipopolysaccharide (LPS).
  • mABs monoclonal antibodies
  • LPS Iipopolysaccharide
  • volumes of both hind paws were measured using a plethysmometer with water cell (12 mm diameter) on Days 0, 5, 7, 10, 14 and 17. Percent inhibition of increase in volume induced by mABs + LPS was calculated by the following formula:
  • Methotrexate a benchmark therapeutic agent, has been shown to yield diminishing benefit in antibody induced models, which are intrinsically less dependent on T-cell activation than on their trafficking and migratory properties.
  • the work of Lange et al. can be cited in this context ⁇ Annals of Rheumatoid Disease 64:599-605, 2005).
  • SymadexTM appears fully active in this model.
  • the results presented in this example are especially relevant to the treatment of human subjects, because the therapeutic effect was obtained by oral administration. In the era of injectable biologies, such as blocking antibodies, the addition of an effective, non-immunosuppressive therapy via the oral route is particularly desirable.
  • Example 7 Symadex " TM downresulates otherwise overexpressed target mechanisms of inflammatory eel! adhesion, cell-surface signaling, and cell proliferation
  • NM_000029 clade A alpha-1 antiproteinase, antitrypsin, member 8
  • CD58 NM_001779 CD58 antigen (lymphocyte function-associated antigen 3)
  • ITSN1 NM_003024 intersectin 1 (SH3 domain protein) potassium inwardly-rectifying channel, subfamily J,
  • KCNJ15 NM_002243 member 15 • potassium voltage-gated channel, KQT-like subfamily,
  • lymphotoxin beta (TNF superfamily, member 3)
  • NCF1 neutrophil cytosolic factor 1 (47kDa, chronic •
  • neutrophil cytosolic factor 2 (65kDa, chronic BI 021745 granulomatous disease, autosomal 1) neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic BI 021745 granulomatous disease, autosomal 1) neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kDa, chronic neutrophil cytosolic factor 2 (65kD
  • VAMP vesicle-associated membrane protein
  • DAVID Database for Annotation, Visualization and Integrated Discovery, from the National Institute of Allergy and Infectious Disease, http://appsl.niaid.nih.gov/david/; the sister program EASE (Expression Analysis Systematic Explorer) at the same site; and the GeneCards bioinformatics project (http://genome- www.stanford.edu/genecards/index.shtml).
  • the down regulated genes ACTA2, ACVRLl, BGN, DSC3, ENG, FBANl 3 FBLNl, HMMR, IGTA2B, ITGA2B, ITGA9, ITGAE, LIMS2, LTB, MAPT, MSLN, NMI, PCDH7, PECAMl, PRDMl, SEMA7A, VAPA all participate in the regulation of these processes via direct modulation of adhesion factors, like integrins and cadherins, or by disrupting the growth factor signals that promote their expression and the assembly of accessory proteins that further facilitate the adhesion process.
  • adhesion factors like integrins and cadherins
  • Neuromodulation via paracrine and autocrine • controls is also evident in the dqwnregulation of systems that further respond to neuroinflammatory insult, including, for example, neurotransmitter transporters associated with damaging, runaway glutamate signaling.
  • the downregu ' lated genes in this latter category are exemplified by ADCYAPl, GABRA3, GGH, KCNQ3, SLCl A2 (and its SLC family solute carrier homologs), and SULT4A1. This latter gene showed close to 300 fold down-regulation. It is a gene associated with heparan sulfation.
  • Sulfated heparans constitute the "molecular velcro" that permits integrins to bind to laminins and thereby provide the linkage that permits invasive inflammatory cells to transmigrate through basal membranes into CNS parenchyma. Down regulation of a such a process would be expected to keep inflammatory cells within the confines of vascular cuffs, as has been observed to be the case in the histopathological evaluation on the SymadexTM treatment effect noted in Examples 2-8.

Abstract

Compounds of formula (I) and a method of treating a patient suffering from an inflammatory and/or demyelinating disorders, comprising administering to said patient a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Definitions for the variables are provided therein.

Description

COMPOUNDS FOR TREATING INFLAMMATORY DISORDERS, DEMYELINATING DISORDERS AND CANCERS
RELATED APPLICATION(S)
This application claims the benefit of U.S. Provisional Application No. 60/771 ,909, filed on February 8/2006 and U.S. Provisional Application No. 60/835,153, filed on August 2, 2006. The entire teachings of the above application(s) are incorporated herein by reference.
BACKGROUND OFTHE INVENTION
There is a need for new pharmaceutically acceptable therapies for various types of cancer as well as inflammatory and demyelinating disorders.
SUMMARY OF THE INVENTION
■The present invention is directed to a class of novel compounds that can be used for treatment of certain cancers, immunocompromising, degenerative, inflammatory disorders and demyelinating diseases, including multiple sclerosis.
In one embodiment, the present invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000002_0001
In formula (I):
R is Rx, a hydrolysable group, or alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group, wherein Rx is -H, an optionally substituted alkyl, hydroxyl, alkoxy group, a halogen, or a group represented by the following structural formula:
Figure imgf000003_0001
or, R and R5 taken together with their intervening carbon atoms form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle; or R and R4 taken together with their intervening carbon atoms form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle;
R2 is -H5 an optionally substituted Cl-ClO alkyl or an optionally substituted aryl or aralkyl or heteroaryl;
R3 is -(CH2)n-NRaRb, wherein n is an integer from 1 to 5, and Ra and Rb, each independently are hydrogen or an optionally substituted alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry, wherein Ry is a heteroaryl or a non-aromatic heterocycle, each optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or Cl- C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl; and
R4, R5 and R6, are each independently -H, -OH5 a halogen or a C1-C6 alkoxy; or
R5 and R6 taken together with their intervening carbon atoms, form a 5, 6 or 7 member, optionally substitited cycloalkyl or non-aromatic heterocycle.
In one embodiment, when R is Rx 5 then Ra and Rb are not H or optionally substituted alkyl and Ry is not optionally substituted N-morpholinyl, optionally substituted N-piperazinyl, or optionally substituted N-pyrazinyl. In another embodiment, when R is Rx, then Ra and Rb are not H or optionally substituted alkyl and Ry is not optionally substituted N-morpholinyl, or optionally substituted N- pyrazinyl.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. IA is a bar plot illustrating inhibition of B-cells proliferation by Symadex™ following stimulation with lipopolysaccharide (LPS). FIG. IB is a bar plot illustrating inhibition of T-cells proliferation- by Symadex™ following stimulation with concavanalin A (Con A).
FIG. 2A is a bar plot illustrating inhibition of IL-4 release by Symadex™ following stimulation with concavanalin A (Con A).
' FIG. 2B is a bar plot illustrating inhibition of IL-10 release by Symadex™ following stimulation with concavanalin A (Con A).
FIG. 3 is a bar plot of the mean clinical score of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ at 20 and 40 mg/kg versus vehicle control.
FIG. 4 is a time dependent chart showing mean clinical score (performance score) of the animals suffering from chronic stage Experimental Autoimmune • Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ versus vehicle control.
FIG. 5 is a time dependent chart showing mean clinical score (performance score) of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 4 weeks of dosing at 20 mg/kg versus vehicle control.
. FIG. 6 is a time dependent chart showing mean clinical score (performance score) of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 6 weeks of dosing at 20 mg/kg versus vehicle control.
FIG. 7 is a time dependent chart showing mean clinical score (performance score) of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 8 weeks of dosing at 20 mg/kg versus vehicle control.
FIG.8 A is a bar chart showing the time course of T-cell counts of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 4, 6 and 8 weeks of dosing at 20 mg/kg versus vehicle control.
FIG.8B is a bar chart showing the time course of CD-4 cell counts of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 4, 6 and 8 weeks of dosing at 20 mg/kg versus vehicle control.
FIG.8C is a bar chart showing the time course of CD-8 cell counts of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 4, 6 and 8 weeks of dosing at 20 mg/kg versus vehicle control.
FIG.8D is a bar chart showing the time course of B-cell counts of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 4, 6 and 8 weeks of dosing at 20 mg/kg versus vehicle control.
FIG. 9 is a time dependent chart showing mean clinical score (performance score) of the animals suffering from chronic stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 2 consecutive doses at 20 mg/kg given 72 hours apart versus vehicle control.
FIG. 10 is a time dependent chart showing mean clinical score of the animals suffering from acute stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 15 consecutive daily dose at 6 mg/kg.
FIG. 11 is a time dependent chart showing mean weight gain record of the animals suffering from acute stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 15 consecutive daily dose at 6 mg/kg.
FIG. 12 is a bar chart showing mean pathology scores on necropsy of τhe animals suffering from acute stage Experimental Autoimmune Encephalomyelitis (EAE) at the indicated day post treatment with Symadex™ after 15 consecutive daily dose at 6 mg/kg.
FIG. 13 is a time dependent chart bar chart showing mean performance of the animals suffering from acute stage Collagen Monoclonal Antibody (mAB) Induced Arthritis at the indicated day post treatment with Symadex™ after 3 consecutive daily oral doses at 30 mg/kg.
FIG. 14 is a list of chemical structures of pharmaceutical agents that can be co-administered with the compounds disclosed in the present invention. DETAILED DESCRIPTION OF THE INVENTION
It has now been discovered that administration of certain imidazoacri dines can be used to treat inflammatory diseases, diseases involving demyelination and certain cancers.
Accordingly, in one embodiment, the present invention is a compound of formula (I) below or a pharmaceutically acceptable salt thereof. In another embodiment, the present invention is a method method of treating an inflammatory or a demyelinating disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
Figure imgf000006_0001
In formula (I), the substituents are each independently defined as follows.
R is Rλ, a hydrolysable group as defined below, or alone or taken together with R4, or alternatively R5, and the intervening carbons is a phenol isosteric group as defined below.
Rx is -H5 alkyl, hydroxy!, alkoxy group, a halogen, or a group represented by the following structural formula:
Figure imgf000006_0002
Included into the group Rx are such values of R that, when R and R5 are taken together with their intervening carbon atoms they form a 5, 6 or 7 member, optionally substituted, cycloaikyl or non-aromatic heterocycle, or such values of R that when R and R4 are taken together with their intervening carbon atoms, they form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle. :
" R2 is -H, an optionally substituted Cl-ClO alkyl or an optionally substituted aryl or heteroaryl.
R3 is -(CH2)n-NRaRb, wherein n is an integer-from 1 to 5, and Ra and Rb, each independently are hydrogen or an optionally substituted alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry.
Ry is a heteroaryl or a non-aromatic heterocycle, each optionally substituted at one or more substitutable carbons with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl.
In one embodiment, when R is Rx, then Ra and Rb are not H or optionally substituted alkyl and Ry is not optionally substituted N-morpholinyl, optionally substituted N-piperazinyl, or optionally substituted N-pyrazinyl.
• In another embodiment, when R is Rx, then Ra and Rb are not H or optionally substituted alkyl and Ry is not optionally substituted N-morpholinyl, or optionally substituted N-pyrazinyl.
When R is Rx such that R and R4 or, alternatively, R and R5 are taken together with their intervening atoms, they preferably form a 5-6 membered cycloalkyl or 5-6 membered non-aromatic heterocycle containing one or two oxygen atoms and optionally substituted with methyl or hydroxyl. More preferably, when R is R\ R4»R5 and R6 are each independently -H, -OH, a C1-C3 alkoxy or taken together with R, forms a methylenedioxy group; and R6 is -H5 -OH, or a C1-C3 alkoxy. More preferably, R4, R5 and R6, are each independently, are independently each -H, -OH, or -OCH3. The remainder of the variables take the values defined above in formula (I).
Preferably, R2 is -H, optionally substituted C1-C8 alkyl, or phenyl, optionally substituted with one or more C1-C4 alkyl, C1-C4 alkoxy, C1-C4 haloalkoxy or cyano groups. More preferably, R2 -H , methyl or ethyl. The remainder of the variables take the values defined above in formula (I). In some embodiments of the compounds of formula (I), Ry is a heteroaryl, preferably N-pyrazinyl or N-pyridinyl. The remainder of the variables take the values defined above in formula (I).
Preferably, n is an integer from 1 to 5, and Ra and Rb, each independently are hydrogen or an alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form a 5-7 membered non-aromatic heterocycle, optionally substituted at one or more substitutable carbons with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein R° and Rd are individually H5 methyl or ethyl.
Preferably, n is 2 or 3 and Ra and Rb, is each independently a hydrogen or a C1-C3 alkyl. Alternatively, Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry. The remainder of the variables take the values defined above in formula (I).
In another preferred embodiment, Ry is a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbons with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR°Rd, wherein Rc and Rd are individually H, methyl or ethyl. -Examples of Ry are listed below:
Figure imgf000008_0001
In these embodiments, Q2 is CH2, NH, or NR102, wherein R102 is methyl or ethyl.
In one embodiment, R is a hydrolysable group. The remainder of the variables take the values defined above in formula (I). As used herein, the term "hydrolysable group" means a derivatives of the compound of formula (I) at position 8, which yields the parent core molecule upon hydrolysis, either spontaneously under acidic or basic conditions in a physiological environment, e.g. blood, metabolically active tissues (liver, kidney, lung, brain) or catalyzed hydrolysis by enzyme, e.g. esterase, peptidases, hydrolases, oxidases, dehydrogenases, lyases or ligases.
Preferably, when R is a hydrolysable group, R is selected from groups represented by structural formulas (II) - (VII):
(HI); "Y R121 (IV);
Figure imgf000009_0002
(γ);
Figure imgf000009_0001
(VI); Q1R10 (VII); R- (VIII); and
Figure imgf000009_0003
(IX),
In variables in structural formulas (II) - (IX) are defined in the paragraphs below.
R7 and R8. are independently each H3 optionally substituted C1-C6 alkyl, optionally substituted aryl or araikyl. Preferably, R7 and R8 are independently each H, optionally substituted C1-C6 alkyl, phenyl or benzyl, optionally substituted with one or more hydroxyl, C1-C3 alkoxy, amino, alkylamino, halogen, C1-C3 haloalkyl or C1-C3 haloalkoxy groups. More preferably, R7 and Rs are each independently H, methyl or ethyl. The remainder of the variables take the values defined above in formula (I).
R9 is carboxyl, carboxamide optionally N-substituted with C1-C4 alkyl, Cl- C6 alkanoyl, C1-C6 carbalkoxy, or optionally substituted aroyl. Preferably, R9 is a C1-C4 alkanoyl. The remainder of the variables take the values defined above in formula (I).
Q1 is O or NH and R10 is H, optionally substituted C1-C6 alkyl or optionally substituted aryl or araikyl. Preferably, R10 is H, C1-C4 alkyl, phenyl or benzyl, optionally substituted with one or more hydroxyl, C1-C3 alkoxy, amino, alkylamino, halogen, haloalkyl or haloalkoxy groups. More preferably, R10 is a H or C1-C4 alkyl. The remainder of the variables take the values defined above in formula (I).
R1 ! and R12 are independently each H, optionally substituted C1-C6 alkyl or, taken together with the atom to which they are attached, form an optionally substituted non-aromatic heterocycle. Preferably, R11 and R12 are independently each a H5 methyl or ethyl or, taken together with the atom to which they are attached form non-aromatic heterocycle, optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl. More preferably, NR11R12 is N-pyrrolidinyl, N-piperidinyl, N-morphoIinyl, N- thiomorpholinyl or N-piperazinyl, optionally N'-substituted with C1-C4 alkyl or Cl- C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H3 methyl or ethyl. The remainder of the variables take the values defined above in formula (I). R13 and R14 are each independently H, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkanoyl, or optionally substituted aroyl, or, taken together with the atom to which they are attached, form an optionally substituted heteroaryl or non-aromatic optionally substituted heterocycle. Preferably, groups R13 and R14 are selected so that group (VI) above is represented by structural formulas (Via) or (VIb):
Figure imgf000010_0001
In structural formulas (Via) and (VIb), Y is a halogen, -NO2, -NH2, -COOH, alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy, and ring A is a 5-7 membered non-aromatic heterocycle optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, oxo or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl. Examples of ring A include the following groups:
Figure imgf000010_0002
Figure imgf000011_0001
The remainder of the variables take the values defined above in formula (I),
R16 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, C1-C6 alkanoyl, or optionally substituted aroyl. Preferably, R16 is optionally substituted C1-C6 alkanoyl; more preferably, R16 is a branched C3-C6 alkanoyl. The remainder of the variables take the values defined above in formula
(I).
R21 is optionally substituted Cl-ClO alkyl, or an optionally substituted aryl or aralkyl or, R21 and R22 taken together with their intervening atoms form a 5-7 membered non-aromatic heterocycle. When R22 is not a part of a heterocycle defined above, the R and R are each independently -H, or optionally substituted C1-C6 alkyl, provided that R22 and R23 are not simultaneously hydrogens. Preferably, either R21 is optionally substituted Cl-ClO alkyl, phenyl or benzyl optionally substituted with a halogen, -NO2.. -NH2, -COOH, alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy, and R22 and R23 are each independently -H, or a C1-C3 alkyl; or R21 and R22, taken together with their intervening atoms, form a 5 or 6 membered non-aromatic heterocycle and R23 is -H, or a Cl -C3 alkyl. The remainder of the variables take the values defined above in formula (I).
R!o° is optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl. In one embodiment, R100 is phenyl or benzyl optionally substituted with a halogen, -NO2, -NH2, -COOH, alkyl, C 2 -C3 "carbalkoxy, C1-C3 alkoxy group, Cl- C3 haloalkyl or C 1 -C3 haloalkoxy. Preferably, R100 is a Cl -C4 alkyl. The remainder of the variables take the values defined above in formula (I).
R101 is H, optionally substituted Cl -C6 alkyl or optionally substituted aryl or aralkyl. In one embodiment, R100 is phenyl or benzyl optionally substituted with a halogen, -NO2, -NH2. -COOH, alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, Cl- C3 haloalkyl or C1-C3 haloalkoxy. Preferably, R!0! is H or C1-C4 alkyl. More preferably, R101 is H5 methyl or ethyl. The remainder of the variables take the values defined above in formula (I).
R107 is C1-C6 alkyl, optionally substituted aryl or aralkyl, or a non -aromatic heterocycle, optionally substituted at one or more substirutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd. Preferably, R107 is C1-C6 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH5 a C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C3 -C3 halbalkoxy or C1-C3 alkyl sulfanyl, or -(CH2)p-(CH2)q-C(O)OH, wherein and q are independently an integer from 1 to 6. More preferably, R107 is C1-C6 alkyl or C1-C6 carboxyalkyl. The remainder of the variables take the values defined above- in formula (I).
In alternative embodiment of the present invention, R alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group. As used herein, the term "phenol isosteric group" means a chemical moiety whose quantum properties result in such electrostatic charge distribution, polarizability, capacity to form hydrogen bonds, hydrophobicity, steric effect and other inductive or mesomeric effects, that the molecule as a whole is endowed with activity as either agonist or antagonist of receptors, enzyme active sites, and protein/enzyme allosteric modulation sites in the context of biological function.
In some embodiments, the phenol isosteric group is selected from the functional groups of formulas (X) - (XXIII):
Figure imgf000012_0001
(XVII); H < (XVIII); (χrχ);
Figure imgf000013_0001
H y- (XXIII).
In formulas (X) - (XXIII), R17 is H3 Cl -C6 alkyl, C1-C6 alkoxyalkyl, optionally substituted aryl or aralkyl or heteroaryl, optionally substituted aryloxy, aralkyloxy or heteroaryloxy, Q is O or S, and Z is CH or N. The remainder of the variables take the values defined above in formula (I).
Preferably R17 is H5 optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxyalkyl, or phenyl, benzyl, phenyloxy or benzylox3', optionally substituted with one or more halogen atoms, -NO2, -NH2, -COOH, C1-C3 alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, Cl -C 3 haloalkyl or C1-C3 haloalkoxy. More preferably, R17 is H, C1-C4 alkyl, or phenyl, optionally substituted with one or more halogen atoms or C1-C3 haloalkyls. Even more preferably, R17 is H, C1-C4 haloalkyl, or phenyl, optionally substituted with one or more C1-C3 haloalkyls. Examples of R17 include H, trifluorom ethyl or phenyl substituted with one or more trifluoromethyls. The remainder of the variables take the values defined above in formula (I).
Examples of compounds of formula (I) include:
Figure imgf000013_0002
Figure imgf000014_0002
(XXVIII);
Figure imgf000014_0001
(XXIX);
Figure imgf000014_0003
The term "alkyl", as used herein, unless otherwise indicated, includes straight or branched saturated monovalent hydrocarbon radicals, typically Cl-ClO, preferably C1-C6. Examples of alkyl groups include, but are not limited to, methyl, " ethyl, propyl, isopropyl, and t-butyl. Suitable substituents for a substituted alkyl include -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy or C1-C3 alkyl sulfanyl, or -(CH2)P- (CH2)q-C(O)OH, where p and q are independently an integer from 1 to 6.
The term "cycloalkyl", as used herein, is a non-aromatic saturated carbocyclic moieties. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. Suitable substituents for a cycloalkyl. are defined above for an alkyl.
The term "haloalkyl", as used herein, includes an alkyl substituted with one or more F, Cl,. Br, or I, wherein alkyl is defined above.
• The terms "alkoxy", as used herein, means an "alkyl-O-" group, wherein alkyl, is defined above.
• The term "alkanoyl", as used herein, means an "alkyl-C(O)-" group, wherein alkyl is defined above.
The term "haloalkoxy", as used herein, means "haloalkyl-O-", wherein haloalkyl is defined above.
. As used herein, an amino group may be a primary (-NH2), secondary (-NHRx), or tertiary (-NRxRy), wherein Rx and Ry may be any of the optionally substituted alkyls alkyls described above.
The term "aryl", as used herein, refers to a carbocyclic aromatic group. Examples of aryl groups include, but are not limited to phenyl and naphthyl.
The term "aryloxy", as used herein, means an "aryl-O-" group, wherein aryl is defined above.
The term "aroyl", as used herein, means an "aryl-C(O)-" group, wherein aryl is defined above.
The term "heteroaryl", as used herein, refers to aromatic groups containing one or more heteroatoms (Θ, S, or N). A heteroaryl group can be monocyclic or polycyclic, e.g. a monocyclic heteroaryl ring fused to one or more carbocyclic aromatic groups or other monocyclic heteroaryi groups. The heteroaryl groups of •this invention can also include ring systems substituted with one or more oxo moieties. Examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, quinolyl, isoquinolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, purinyl, oxadiazolyl, thiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, dihydroquinolyl, tetrahydroquinolyl, dihydroisoquinolyl, tetrahydroisoquinolyl, benzofuryl, furopyridinyl, pyrolopyrimidinyl, and azaindolyl.
The term "heteroaryloxy", as used herein, means a "heteroaryl-O-" group, wherein heteroaryl is defined above.
The term "non-aromatic heterocycle" refers to non-aromatic carbocyclic ring systems typically having four to eight members, preferably five to six, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N3 O, or S. Non aromatic heteroccyles can be optionally unsaturated. Examples of non-aromatic heterocyclic rings include 3-tetrahydrofuranyl, 2-tetrahydropyranyl, 3-tetrahydropyranyl, 4-tetrahydropyranyl, [l,3]-dioxalanyl, [l,3]-dithiolanyl, [1,3]- dioxanyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3- morpholinyl, 4-morpholinyl, 2-thiomorphoIinyl, 3-thiomorpholinyl, 4- thiomorpholinyl, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3-pyrorolidinyl, 1 -piperazinyl, 2- piperazinyl, 1 -piperidinyl, 2-ρiρeridinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, N-substituted diazolonyl, and 1-pthalimidinyl.
The heteroaryl or non-aromatic heterocyclic groups may be C-attached or N- attached (where such is possible). For instance, a group derived from pyrrole may be pyrrol-1-yl (N-attached) or pyrrol-3-yl (C-attached).
Suitable substituents for an aryl, a heteroaryl, or a non-aromatic heterocyclic group are those that do not substantially interfere with the pharmaceutical activity of the disclosed compound. One or more substituents can be present, which can be identical or different. Examples of suitable substituents for a substitutable carbon atom in aryl, heteroaryl or a non-aromatic heterocyclic group include -OH, halogen (-F, -Cl, -Br5 and -I), -R', haloalkyl, -OR5, -CH2R', -CH2OR', -CH2CH2OR', -CH2OC(O)R', -O-COR', -COR', -SR', -SCH2R', - CH2SR', -SOR', -SO2R', -CN5 -NO23 -COOH, -SO3H3 -NH2, -NHR', -N(R')2, -COOR', -CH2COOR', -CH2CH2COOR', -CHO, -CONH2, -CONHR', -CON(R')2, -NHCOR' , -NR' COR', -NHCONH2, -NHCONRΗ, -NHCON(R')2, -NR5CONH23 -NR'CONR'H, -NR'CON(R')2, -C(=NH)-NH2, -C(=NH)-NHR\ -C(=NH)-N(R')2, -C(=NR')-NH2s -C(=NR')-NHR\ -C(=NR')-N(R')2, -NH-C(=NH)-NH2, -NH-C(=NH)-NHR\ -NH-C(=NH)-N(R')2, -NH-C(=NR')-NH2, -NH-C(=NR')-NHR\ -NH-C(=NR')-N(R')2, -NR'H-C(=NH)-NH2, -NR'-C(=NH)-NHR', -NR'-C(=NH)-N(R')2, -NR'-C(=NR')-NH2, -NR'-C(=NR')-NHR\ -NR'-C(=NR')-N(R')2, -SO2NH2, -SO2NHR', -SO2NR'2, -SH5 -SOkR' (k is 0, 1 or 2) and -NH-C(=NH)-NH2. Each R' is independently an alkyl group. Oxo (C=O) and thio (C=S) are also suitable substituents for a non-aromatic heterocycle.
Suitable substituents ori the nitrogen of a non-aromatic heterocyclic group or a heteroaryl group include -R%", -N(R")2, -C(O)R", -CO2 R", -C(O)C(O)R", -C(O)CH2 C(O)R", -SO2R", -SO2 N(R")2, -C(=S)N(R")2, -C(=NH)-N(R")2, and -NR" SO2R". R" is hydrogen, an alkyl or alkoxy group.
Compounds of formula (I) can be synthesized according to a variety of synthetic schemes disclosed in U.S. Pat. Nos. 5,231,100 and 6,229,015, incorporated herein by reference in their entirety. One example of such a scheme is shown below:
Figure imgf000017_0001
(Scheme I)
As used herein, the "activated alkanoic acylating agent" is defined within the references cited. For example: alkyl nitriles are reacted with HCl in methanol or ethanol to give the corresponding acetimidate ester hydrochlorides, R-CN going to R-C(OMe)=NH+ Cl-, where R is an alkyl. This acetimidate reacts with the amine in compound (S 1.3) and cyclized to the methyl or ethyl or other alkylimidazole, again per the cited articles.
It has now been discovered, that Symadex™ inhibits proliferation of B-cells following stimulation with LPS and T-cells following stimulation with Con A as well as that Sytnadex™ inhibit release of cytokines such as IL-4 and IL-IO (Example 1). It has further been discovered in microarray experiments, that Symadex™ treatment results in altered expression of several genes involved in key regulatory pathways affecting the inflammatory and proliferative states, particularly " the ability of invasive cells to assemble and aggregate,- downregulation of cell proliferation and cell-cell signaling (Example 3). These molecular pharmacology studies show that Symadex™ exerts a downregulatory effect on genes implicated in mechanisms of cell aggregation and proliferation and on processes associated with invasive cellular growth, which are the hallmark of the inflammatory etiology associated with the autoimmune diseases. Taken together, these results indicate that Symadex™ can be used for treating the disorders that have inflammatory component, including autoimmune diseases.
It was further discovered that Symadex™ demonstrates activity in the female Hartley guinea pig Experimental Autoimmune Encephalomyelitis (EAE) model, a classic animal model for chronic-progressive MS (Example 2). Taken together with the results of Example 3, this result indicates that Symadex™ can be used for treating the disorders that have demyelinating as well as inflammatory components.
Accordingly, in one embodiment, the present invention is a method of treating a patient suffering from an inflammatory condition. The condition can be systemic lupus, inflammatory bowl disease, psoriasis, Crohn's disease, rheumatoid arthritis, sarcoid, Alzheimer's disease, a chronic inflammatory demyelinating neuropathy, insulin dependent diabetes mellitus, atherosclerosis, asthma, spinal cord injury or stroke.
Examples of chronic inflammatory demyelinating neuropathies include: chronic Immune Demyelinating Polyneuropathy (CIDP); multifocal CIDP; multifocal motor neuropathy (MMN); anti-MAG Syndrome (Neuropathy with IgM binding to Myelin-Associated Glycoprotein); GALOP Syndrome (Gait disorder Autoantibody Late-age Onset Polyneuropathy); anti-sulfatide antibody syndrome; anti-GM2 gangliosides antibody syndrome; POEMS syndrome (Polyneuropathy Organomegaly Endocrinopathy or Edema M-protein Skin changes); perineuritis; and IgM anti-GDlb ganglioside antibody syndrome. The method comprises administering to a patient a therapeutically effective amount of a compounds of formula (I) or a pharmaceutically acceptable salt thereof.-
In another embodiment, the present invention is a method of treatment of a patient suffering from a demyelinating condition. As used herein,- a "demyelinating condition" is a condition that destroys, breaks the integrity of or damages a myelin sheath. As used herein, the term "myelin sheath" refers to an insulating layer ' surrounding vertebrate peripheral neurons, that increases the speed of conduction and formed by Schwann cells in- the peripheral or by oligodendrocytes in the central nervous system. Such condition can be multiple sclerosis, a congenital metabolic disorder, a neuropathy with abnormal rήyelination, drug-induced demyelination, radiation induced demyelination, a hereditary demyelination condition, a prion- induced demyelination, encephalitis-induced demyelination, a spinal cord injury, Alzheimer's disease as well as chronic inflammatory demyelinating neuropathies, examples of which are given above. In one embodiment; the condition is multiple sclerosis. The method comprises administering to a patient a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
The term "patient" means a warm blooded animal, such as for example rat, mice, dogs, cats, guinea pigs, and primates such as humans. The terms "treat" or "treating" include any treatment, including, but not limited to, alleviating symptoms, eliminating the causation of the symptoms either on a temporary or permanent basis, or preventing or slowing the appearance of symptoms and progression of the named disorder or condition. The term "therapeutically effective amount" means an amount of the compound, which is effective in treating the named disorder or condition. In certain embodiments, therapeutically effective amount means an amount sufficient to effect remyelination of nerve cells in a patient.
In another embodiment, the present invention is a method of promoting remyelination of nerve cells in a patient, comprising administering to the patient in need thereof a therapeutically effective amount of a compound of formula (I). The patient can be suffering from any of the demyelinating conditions listed above.
In another embodiment, the present invention is a method of preventing demyelination and promoting remyelination in a patient in need thereof, comprising administering a combination of a therapeutically effective amount of a compound of formula (I) or pharmaceutically acceptable salt thereof, and an anti-inflammatory agent as" described below.
In another embodiment, the present invention is a method of reversing paralysis in a subject in need thereof with a demyeliriating disease, comprising administering to the subject a compound in an amount sufficient to inhibit lymphocyte infiltration of immune cells in the spinal cord to promote remyelination of nerve cells in the spinal cord and .thereby treating paralysis in said subject, wherein the compound is of formula formula (I) or a pharmaceutically acceptable salt thereof.
. The dosage range at which the disclosed .compounds of formula (I) exhibit their ability to act therapeutically can vary depending upon the severity of the condition, the patient, the formulation, other underlying disease states that the patie.nt is suffering from, and other medications that may be concurrently administered to the patient. Generally, the inventive compounds of the invention will exhibit their therapeutic activities at dosages of between about 0.001 mg/kg of patient body weight/day to about 100 mg/kg of patient body weight/day. For example, the dosage can be 0.1-100 mg/kg, 1 -100 mg/kg, 10-100 mg/kg, 1-50 mg, kg,.10-50 mg/kg or 10-30 mg/kg per day, per every other day or per week.
In other embodiments, compounds can be administered by any of the routes described below, preferably intravenously, in an amount from 1 mg per kilogram, body weight to 20 mg per kg body weight. Compounds can be administered daily, once every 72 hours or weekly.
In one embodiment in which compounds are used to treat rheumatoid arthritis; compounds can be administered orally in an amount of 1-50 mg/kg, 10-40 mg/kg, 20-30 mg/kg or 30 mg per kilogram of body weight per day, per every other day or per week.
In one embodiment, the compounds of the invention are administered chronically to the patient in need thereof. For example, the chronic administration of the compound is daily, weekly, biweekly, or monthly over a period of at least one year, at least two years, at least three or more years. In one embodiment, the compounds of formula (I) axe administered intravenously in the amount of 1.5-30 mg/kg once at intervals of 1-3 months. In another embodiment, the compounds are administered orally in the amount of 5-100 mg/kg on same schedule as above. Alternatively, the compounds of formula (I) are administered several times over a period of up to 3 months and up to a cumulative dose of between 1.5 and 30 mg/kg. In another embodiment, the cumulative dose is from 5 to 100 rng/kg.
In another embodiment, the compounds of formula (I) are administered intravenously in the amount of 2.5-10 mg/kg weekly for 8-24 weeks, repeating as needed after 6-18 weeks off drug. Alternatively, the compounds of formula (I) are administered several times over a period of from 14 weeks to 42 weeks to achieve a cumulative dose from 20 mg/kg to 240 mg/kg. Administration can be repeated over one or more periods of 14-42 weeks.
In another embodiment, the compounds of formula (I) are administered intravenously in the amount of 2.5-10 mg/kg twice, 72 hrs apart for 1 to 2 weeks, repeating monthly. Alternatively, the compounds of formula (I) are administered several times over a period of up to two weeks, up to a cumulative dose of from 11 mg/kg to 47 mg/kg. Administration can be repeated monthly.
In another embodiment, the compounds of formula (I) are administered orally in the amount of 1-3 mg/kg daily for 10-15 days, repeating every 30-45 days. Alternatively, the compounds of formula (I) are administered several times over a period of up to 40-60 days, up to a cumulative dose of from 10 mg/kg to 45 mg/kg. Administration can be repeated over one or more periods of up to 40-60 days.
In another embodiment, the compounds of the invention are administered orally in the amount of 2-6 mg/kg daily for 3 days per week, repeating every 15-30 days. Alternatively, the compounds of formula (I) are administered several times over a period of up to 30 days up to a cumulative dose of 6-18 mg/kg. Administration can be repeated over one or more periods of up to 30 days.
Preferably, the administration of the compounds or the combinations of the compounds described herein results in an effective blood level of the compound in the patient of more than or equal to 10 ng/ml. For example, compounds can be administered intravenously in an amount of 20 μg to about 500 μg per kilogram body weight of the patient.
Preferred human doses for treating chronic (remitting-relapsing) multiple sclerosis (MS) are 0.1 mg/kg to 10 mg/kg, 1-10 mg/kg,- 1-5 mg/kg, 2-7'mg/kg, 2 - 5 mg/kg. Schedule could be once a month, twice a month, three times a month or once or twice a week for 3 months, 6 month, 12 months or more. ' .
Preferred human doses for treating acute MS, is 0.1 mg/kg to 10 mg/kg, 0.1 - 5 mg/kg, 0.1 -2 mg/kg, 0.5-2 mg/kg or 0.5 - 1 mg/kg three times a day, twice a day, or daily, on a weekly, biweekly or monthly basis.
Preferred human doses for treating rheumatoid arthritis 0.1 mg/kg to 10 mg/kg, 1-10 mg/kg, 1-5 mg/kg, 2-7 mg/kg, 2 - 5 mg/kg three times a day, twice a day, or daily, on a weekly, biweekly or monthly basis.
The compounds used in the present invention can be administered alone or in combination with one or more other pharmaceutically active agents that are' effective against the inflammatory condition and/or the demyelating disorder being treated.
As used herein, the term "combination" with reference to pharmaceutically active agents and the term "co-administering" and "co-administration" refer to administering more than one pharmaceutically active agent to a patient during one- treatment cycle and not necessarily simultaneous or in a mixture.
In one embodiment, the compounds of the present invention are administered in combination- with an anti-inflammatory agent. The anti-inflammatory agent can be adrenocorticotropic hormone, a corticosteroid, an interferon, glatiramer acetate, or a non-steroidal anti -inflammatory drug (NSAID).
Examples of suitable anti -inflammatory agents include corticosteroid such as prednisone, methylprednisolone, dexamethasone Cortisol, cortisone, fludrocortisone, prednisolone, 6α-methylprednisolone, triamcinolone, or betamethasone.
Other examples of suitable anti -inflammatory agents include NSAIDs such as aminoarylcarboxylic acid derivatives (e.g., Enfenamic Acid, Etofenamate, Flufenamic Acid, Isonixin, Meclofenamic Acid, Nifiumic Acid, Talniflumate, Terofenamate and Tolfenamic Acid), arylacetic acid derivatives (e.g., Acematicin, Alclofenac, Amfenac, Bufexamac, Caprofen, Cinmetacin, Clopirac, Diclofenac, Diclofenac Sodium, Etodolac, Felbinac, Fenclofenac, Fenclorac, Fenclozic Acid, Fenoprofen, Fentiazac, Flubiprofen, Glucametacin, Ibufenac, Ibuprofen, Indomethacin, Isofezolac, Isoxepac, Ketoprofen, LonazolaC, Metiazinic Acid, Naproxen, Oxametacine, Proglumrtacin, Sulindac, Tenidap, Tiramide, Tolectin, Tolmetin, Zomax and Zomepirac), arylbutyric acid ferivatives (e.g., Bumadizon, Butibufen, Fenbufen and Xenbucin) arylcarboxylic acids (e.g., Clidanac, Ketorolac and Tinoridine), arylproprionic acid derivatives (e.g., Alminoprofen, Benoxaprofen, Bucloxic Acid, Carprofen, Fenoprofen, Flunoxaprofen, Flurbiprofen, Ibuprofen, Ibuproxam, Indoprofen, Ketoprofen, Loxoprofen, Miroprofen, Naproxen, Oxaprozin, Piketoprofen, Piroprofen, Pranoprofen, Protinizinic Acid, Suprofen and Tiaprofenic Acid), pyrazoles (e.g., Difenamizole and Epirizole), pyrazolones (e.g., Apazone, Benzpiperylon, Feprazone, Mofebutazone, Morazone, Oxyphenbutazone, Phenylbutazone, Pipebuzone, Propyphenazone, Ramifenazone, Suxibuzone and Thiazolinobutazone), salicyclic acid derivatives (e.g., Acetaminosalol, 5- Aminosalicylic Acid, Aspirin, Benorylate, Biphenyl Aspirin, Bromosaligenin, Calcium Acetylsalicylate, Diflunisal, Etersalate, Fendosal, Flufenisal, Gentisic Acid, Glycol Salicylate, Imidazole Salicylate, Lysine Acetylsalicylate, Mesalamine, Morpholine Salicylate, 1-Naphthyl Sallicylate, Olsalazine, Parsalmide, Phenyl Acetylsalicylate, Phenyl Salicylate, 2-Phosphonoxybenzoic Acid, Salacetamide, Salicylamide O-Acetic Acid, Salicylic Acid, Salicyloyl Salicylic Acid, Salicylsulfuric Acid, Salsalate and Sulfasalazine), thiazinecarboxamides (e.g., Droxicam, Isoxicam, Piroxicam and Tenoxicam), e-Acetamidocaproic Acid, S- Adenosylmethionine, 3-Amino-4-hydroxybutyric Acid, Amixetrine, Bendazac, Benzydarnine, Bucolome, Difenpiramide, Ditazol, Emorfazone, Guaiazulene, Ketorolac, Meclofenamic Acid, Mefenamic Acid, Nabumetone, Nimesulide, Orgotein, Oxaceprol, Paranyline, Perisoxal, Pifoxime, Piroxicam, Proquazone, Tenidap and a COX-2 inhibitor (e.g., Rofecoxib, Vaidecoxib and Celecoxib).
Further examples of anti -inflammatory agents include aspirin, a sodium salicylate, choline magnesium trisalicylate, salsalate, difiunisal, sulfasalazine, olsalazine, a para-aminophenol derivatives, an indole, an indene acetic acid, a heteroaryl acetic acid, an anthranilic acid, an enolic acid, an alkanones, a diaryl- substituted furanone, a diaryl-substituted pyrazoles, an indole acetic acids, or a sulfonanilide. In some embodiments, the compounds of the present invention can be administered in combination with immunotherapeutic agents such as interferons and anti-integrin blocking antibodies like natalizumab.
Examples of agents suitable for treating demyelinating disorders include Pirfenidone, Epalrestat, Nefazodone hydrochloride, Memantine hydrochloride, Mitoxantrone hydrochloride, Mitozantrone hydrochloride, Thalidomide, Roquinimex, Venlafaxine hydrochloride, Intaxel, Paclitaxel, recombinant human nerve growth factor: nerve growth factor, ibudilast, Cladribine, Beraprost sodium, Levacecarnine hydrochloride, Acetyl-L-carnitine hydrochloride; Levocarnitine acetyl hydrochloride, Droxidopa, interferon alfa, natural interferon alpha, human lymphoblastoid interferon, interferon beta-lb, interferon beta-Ser, Alemtuzumab, Mycophenolate mofetil, Zoledronic acid monohydrate, Adapalene, Eliprodil, Donepezil hydrochloride, Dexanabinol, Dexanabinone, Xaliproden hydrochloride, interferon alfa-n3, lipoic acid, thioctic acid, Teriflunomide, Atorvastatin, Pymadin, 4-Aminopyridine, Fampridine, Fidarestat, Priliximab, Pixantrone maleate, Dacliximab, Daclizumab, Glatiramer acetate, Rituximab, Fingolimod hydrochloride, interferon beta-la, Natalizumab, Abatacept, Temsirolimus, Lenercept, Ruboxistaurin mesilate hydrate, Dextromethorphan/quinidine sulfate, Capsaicin, Dimethylfumarate or Dronabinol/cannabidiol.
In some embodiments, the compounds of the present invention can be administered in combination with one or more other pharmaceutically active agents that are effective against multiple sclerosis. Examples of such agents include the interferons (interferon beta 1-a, beta 1-b, and alpha), gϊatiramer acetate or corticosteroids such as methylprednisolone and prednisone as well as chemotherapeutic agents such as mitoxantrone, methotrexate, azathioprine, cladribine cyclophosphamide, cyclosporine and tysabri.
Further examples of pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include compounds of the following structural formulae:
Figure imgf000025_0001
.
Figure imgf000025_0002
Figure imgf000026_0001
Figure imgf000026_0002
Figure imgf000027_0001
Figure imgf000027_0002
Figure imgf000027_0003
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000029_0002
Further examples of pharmaceutical agents that can be co-administered with the compounds of formula (I) include:
T-cell receptor (TCR) Vβ6 CDR2 peptide vaccine consisting of TCR Vβ6, amino acid sequence 39-58, Leu - GIy - GIn - GIy - Pro - GIu - Phe - Leu - Thr - Tyr
- Phe - GIn - Asn - GIu - Ala - GIn - Leu - GIu - Lys - Ser (SEQ ID NO: 1);
Myelin basic protein immunogen peptide, aminoacid sequence 75-95, Lys - Ser - His - GIy - Arg - Thr - GIn - Asp - GIu - Asn - Pro - VaI - VaI - His - Phe - Phe
- Lys - Asn - He - VaI - Thr (SEQ ID NO:2);
Tiplimotide, myelin basic protein immunogen vaccine peptide, aminoacid sequence 83-99, D - Ala - lys - pro - val - val - his - leu - phe - ala - asp - ile - val - thr - pro - arg - thr - pro, (SEQ ID NO:3);
Myelin basic protein immunogen peptide, aminoacid sequence 82-98, Asp - glu - asp - pro - val - val - his - phe - phe - lys - asp - ile - val - thr - pro - arg - thr, (SEQ ID NO:4);
Adrenocorticotropic hormone (ACTH), Ser - Tyr - Ser - met - glu - his - phe
- arg - try - gly - lys - pro - val - gly - lys- lys - arg - arg - pro - val - lys - val - tyr- pro - asp - gly - ala - glu - asp - glu - leu - ala - glu - ala - phe - pro - leu - glut - phe, (SEQ ID NO:5).
Further examples of pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include: 3-4 diaminopyridine; ABT-874; Actos® (pioglitazone); ALCAR (acetyl-L- carnitine); Alpha lipoic acid; AndroGel® (testosterone gel); combination of trimethoprim and vitamin C; combination of azithromycin and rifampin; minocycline; donezepil HCL; Avandia® (rosiglitazone maleate; combination of IFN beta-la) and acetaminophen, ibuprofen or prednisone; combination of Avonex® (interferon beta- Ia) + CellCept®
(mycophenolate mofetil); combination of Avonex® (interferon beta- Ia) and Copaxone® (glatiramer acetate); combination of Avonex® (interferon beta- Ia) and doxycyciine; combination of Avonex® (interferon beta-1 a) and EMLA (lidocaine and prilocaine) anesthetic cream; Avonex® (interferon beta-la) and estrogen and progesterone; combination of Avonex® (interferon beta-1 a) + Fludara® (fludarabine phosphate); combination of Avonex® (interferon beta- Ia) and methotrexate and leucovorin rescue; combination of Avonex® (interferon beta- Ia) and methotrexate and methylprednisolone; combination of Avonex® (interferon beta- Ia) and Novantrone® (mitoxantrone); combination of Avonex® (interferon beta-la) and Prozac® (fluoxetine); combination of Avonex® (interferon beta-la) and Topamax® (topiramate); combination of Avonex® (interferon beta- Ia) and Zocor® (simvastatin); AVP-923 (dextromethorphan/quinidine); combination of Betaseron® (interferon beta-1 b) and Imuran® (azathioprine); combination of Betaseron® (interferon beta-1 b) and Copaxone® (glatiramer acetate); combination of BHT-3009- 01 and Lipitor® (atorvastatin); Bone marrow/peripheral stem cell transplant; CellCept® (mycophenolate mofetil)'; combination of CellCept® (mycophenolate mofetil) and Avonex® (interferon beta-la); Oral ciadribine; CNTO 1275 (monoclonal antibody); combination of Copaxone® (glatiramer acetate) and Antibiotic therapy (minocycline); combination of Copaxone® (glatiramer acetate) and Novantrone® (mitoxantrone); combination of Copaxone® (glatiramer acetate) and prednisone; combination of Copaxone® (glatiramer acetate) and Proventil® (albuterol); Cyclophosphamide; Daclizumab; Deskar® (pirfenidone); Estriol; Fumaric acid esters; Gabitril® (tiagabine HCL); Ginkgo biloba; IDEC-131 (anti- CD40L or anti-CD 154); the combination of Immunoglobulin and methylprednisolone; Inosine; Interferon tau; Lamictal® (lamotrigme); Lexapro® (escitalopram); Lipitor® (atorvastatin); combination of Lipitor® (atorvastatin) and Rebif® (interferon beta- Ia); combination of Lymphocytapheresis (removal of immune cells), Imuran® (azathioprine) and prednisone; MBP8298; Methylprednisolone; combination of Methylprednisolone and Avonex (interferon beta-la); Modiodal (modafmil); NBI- 5788 (altered peptide ligand); combination of Novantrone® (mitoxantrone for injection concentrate) and Avonex® (Interferon beta-la) or Copaxone® (glatiramer acetate); Omega-3 Fatty Acid Supplementation; Pixantrone (BBR 2778); combination of Provigil® (modafinil) and Avonex® (interferon beta- Ia); Rapamune® (sirolimus); RG2077; Rituxan® (rituximab); Rolipram (phosphodiesterase-4 inhibitor); SAIK-MS (laquinimod, ABR-215062); T cell vaccination; Teriflunomide; Tetrahydrocannabinol; Tetrahydrocannabinol (dronabinol); Thalamic stimulation; combination of Tysabri® (natalizumab) and Avonex® (interferon beta- Ia); combination of Tysabri® (natalizumab) and Copaxone® (glatiramer acetate); and Viagra® (sildafenil citrate).
Further examples of pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include compounds listed in FIG. 14. Additionally, Copaxone (Glatiramer) can be orally co-administered with the compounds of the present invention.
In other embodiments, pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include compounds include: Mylinax, an oral formulation of cladrlbine used in leukaemia treatment, developed by Serono/Ivex; Teriflunomide. a metabolite of Arava, an oral immunosuppressant, developed by Sanofl-Aventis; FTY 720, an oral immunomodulator (Sphingosine-1 -phosphate receptor agonist), developed by Novartis; MBP 8298, a synthetic myelin basis protein designed to reduce the emergence of antibodies directed against the myelin, developed by Bio MS Medical; an orphan drug 4-aminopyridline (4-AP), a potassium channel blocker, developed by Acorda; Gamunex, an intravenous immunoglobulin formulation, developed by Bayer; BG-12 fumarate, a second generation oral futnarate, developed by Biogen Idec/Fumapharm; Temsirolirnus, a T-lymphocytes proliferation blocker, developed by Wyeth; E-2007, an AMPA receptor agonist, developed by Eisal; Campath, a humanized antibody directed against CD52, developed by'Genzyme; Neuro V ax, a vaccine, developed by Immune Response; Zocor, a statin, developed by Merck; NBI 5788, a myelin- mimicking peptide Hgand; developed by Neurocrine; Tauferon, Interferon tau, developed4 by Pepgen; Zenapax, a humanized anti-CD25 immunosuppressive antibody, developed by Protein Design; a combination of MS-EET and EMZ 701, a . methyl donator, developed by Transition Therapeutics; Laquinlmod, an oral formulation of a derivative of linomide, developed by Active Biotecb/Teva; deskar pirfenidone^ a TNF -alpha inhibitor, developed by Mamac; ATL-1102, a second generation antisense inhibitor targeting VLA4, developed by Antisense Therapeutics. . •
In some embodiments, compounds of formula (A) can be administered in combination with antivascular agents, in particular agents inhibiting the growth " factor receptors, Epidermal Growth Factor Receptor. (EGFR), Vascular Epidermal Growth Factor Receptor (VEGFR), and Fibroblast Growth Factor Receptor (FGFR). Examples of such agents include, Iressa, Tarceva, Erbitux, Pelitinib, AEE-788, CP- 547632, CP-547623,-Tykerb (GW-2016), INCB-7839, ARR Y-334543, BMS- 599626, BIBW-2992, Falnidamol, AG1517, E-7080, KRN-951, GFKI-258, BAY- 579352, CP -7055, CEP-5214, Sutent, Macugen, Nexavar, Neovastat, Vatalanib succinate, GW-78603413, Lucentis, Teavigo, AG-13958, AMG-706, Axitinib; " • ABT-869, Evizon, Aplidin, NM-3, PI-88, Coprexa, A2D-2171, XL-189, XL-880, XL-820, XL-647, ZK-CDK, VEGFTrap, OSI-930, Avastin, Revlimid, Endostar, Linomide, Xinlay, SU-όόS, BIBF-Ϊ 120, BMS-5826624, BMS-540215.
In some embodiments, compounds of formula (I) can be administered in combination with agents that affect T-cell homing, extravastion and transmigration. Examples of such agents include, FTY~720PKI-166, PTK-787, SU-11248.
In some embodiments, compounds of formula (I) can be administered in combination with agents inhibiting VLA-4. Examples of such agents include, Tysabri, Bio-121 1. HMR- 1031, SB-683698, RBx-4638,RO-0272441, RBx- 7796,SB-683699, DW-908e, AJM-300, and PS-460644.
Daily dose of administration of the compounds of the present invention can be repeated, in one embodiment, for one week. In other embodiments, daily dose can be repeated for one month to six months; for six months to one year; for one year to five years; and for five years to ten years. In other embodiments, the length of the treatment by repeated administration is determined by a physician.
In another embodiment, the present invention is a method of treating a subject suffering from, a cancer. As used herein,' the term "cancer" refers to the uncontrolled growth of abnormal cells that have mutated from normal tissues. A cancerous tumor (malignancy) is of potentially unlimited growth and expands locally by invasion and systemically by metastasis. Examples of cancers that can be treated by the compounds of the present invention include: colorectal cancer, non- small cell lung cancer, ovarian, renal, sarcoma, melanoma, head & neck, hepatocellular, thyroid, multidrug-resistant leukemia, lymphoma, and multiple myeloma.
• "Treating a subject suffering from cancer" includes achieving, partially or substantially, one or more of the following: arresting the growth or spread of a cancer, reducing the extent of a cancer (e.g., reducing size of a tumor or reducing the number of affected sites), inhibiting the' growth rate of a cancer, and ameliorating or improving a clinical symptom or indicator associated with a cancer (such as tissue or serum components).
Treating, as used herein, refers to partially or totally inhibiting, delaying, or reducing (partially or completely) the progression of cancer including cancer metastasis; inhibiting, delaying or preventing the recurrence of cancer including cancer metastasis in,. for example, a human.
In one embodiment the cancer selected from the group consisting of ovarian cancer, melanoma, renal cancer, sarcoma (e.g., liposarcoma and leiomyosarcoma), hepatocellular cancer, thyroid cancer, head and neck cancer, non-small cell lung cancer and colorectal cancer is a solid tumor. As used herein, a "solid tumor" is, used to distinguish between a localized mass of tissue and leukemia. Leukemia is a cancer that starts in blood-forming cells of the bone marrow as a result of an abnormal development of leukocytes (white blood cells) and their precursors. .
Different kinds of solid tumors are named for the type of cells of which they are composed and include but are not limited to, for example, sarcomas (cancers arising from connective or supporting tissues such as bone or muscle), carcinomas (cancers arising from the body's glandular cells and epithelial cells, which line body tissues) and lymphomas (cancers of the lymphoid organs such as the lymph nodes, spleen, and thymus, which produce and store infection-fighting cells).
As used herein the term "cancer" can be primary or secondary. Primary cancer is named after their original cell type, from the organ where the cancer first begins to grow. Cancer cells can break away from the primary site and travel to other parts of the body in the blood or lymphatic system. The cells eventually lodge in another body organ and begin to grow there. This is called a secondary cancer.
As used herein "ovarian cancer", is cancer of the ovaries or fallopian tubes, including cancers of germ cells, stromal cells, and epithelial cells. Examples of ovarian cancers include but are not limited to:
Epithelial Ovarian Tumors, which include but are not limited to, serous adenomas, mucinous adenomas, and Brenner tumors, rumors of low malignant potential (LMP tumors), borderline epithelial ovarian cancer, epithelial ovarian cancers, carcinomas and undifferentiated epithelial ovarian carcinomas;
Germ Cell tumors which include but are not limited to, teratoma, dysgerminoma, endodeππal sinus tumor, and choriocarcinoma; and
Stromal tumors, which include but are not limited to, granulosa cell tumors, granulosa-theca tumors, and Sertoli-Leydig cell tumors.
"Renal cancer" or "kidney cancer", as used herein, includes but is not limited to, transitional cell cancer (TCC) of the renal pelvis, Wilms Tumour and renal cell cancer.
Renal cell cancer is also called renal adenocarcinoma or hypernephroma. In renal cell cancer, the cancerous cells are found in the lining of the tubules (the smallestjubes inside the nephrons that help filter the blood and make urine).
There are several types of renal cell cancer including but not limited to clear cell, chromophilic, chromophobic, oncocytic, collecting duct and sarcomatoid.
Renal cancer also includes cancers containing more than one of the cell types described above.
As used herein, "melanoma" is a type of skin cancer that occurs in the cells that color the skin, called melanocytes. Types of melanoma include but are not limited to: Cutaneous melanoma, superficially spreading melanoma, nodular malignant melanoma, lentiginous malignant melanoma, acral lentiginous melanoma, demoplastic malignant melanomas', giant melanocyte nevus, amelandtic malignant melanoma, acral lentiginous melanoma unusual melanoma variants, including mucosal malignant melanoma and ocular malignant 'melanoma.
"Sarcomas", as used herein, include but are not limited to, fibrosarcomas from fibrous body tissues, leiomyosarcomas and rhabdomyosarcomas from muscle tissues, liposarcomas from fat, synovial sarcomas, angiosarcomas from blood vessels, MPNST - malignant peripheral nerve sheath tumours (PNSTs), GIST - gastrointestinal stromal sarcoma, osteosarcoma, myosarcoma, chondrosarcoma, bile duct sarcoma, brain sarcoma, breast sarcoma, soft tissue sarcoma, uterine sarcoma, endocardial sarcoma, stromal sarcomas from supporting tissues (endometrial stromal sarcoma), granuloytic, histiolytic, hemangioendothelial, Kupffer-cell, neurogenic, round-cell, reticulum cell, spindle cell, Kaposi's sarcoma of the skin, Ewing's sarcomas and PNETs. In certain embodiments, the sarcoma is leiomyosarcoma or liposarcoma.
"Thyroid cancer", as used herein, includes but is not limited to, papillary and/or mixed papillary/follicular, follicular and/or Hurthle cell, lymphoma, medullary, anaplastic and combinations thereof.
The term "head and neck cancer" as used herein, encompasses tumors that occur in several areas of the head and neck region, including the nasal passages, sinuses, mouth, throat, larynx (voice box), swallowing passages, salivary glands, and skin cancers that develop on the scalp, face, or neck may also be considered head and neck cancers. These cancers include but are not limited to squamous cell carcinoma, mucoepidermoid carcinoma, adenoid cystic carcinoma, lymphoma, adenocarcinoma, esthesioneuroblastoma, tumors of the nasal cavity and paranasal sinuses, nasopharyngeal cancer, cancers of the oral cavity (including all the various parts of the mouth: the lips; the lining inside the lips and cheeks (the buccal mucosa); the bottom of the mouth; the front of the tongue; the front part of the top of the mouth (the hard palate); the gums; and the area behind the wisdom teeth (the retromolar trigone)), tumors of the oropharynx, hypopharyngeal tumors, laryngeal cancer and salivary gland cancer (including malignant salivary gland tumor). As used herein "hepatocellular cancer" or "liver cancer" includes but is not limited to: hepatocellular carcinoma (also sometimes called hepatoma or HCC) "carcinoma", fibrolamellar HCC5 cholangiocarcinoma, angiosarcoma (also be called haemangiosarcoma) and hepatoblastoma.
As used herein, "non-small, cell lung cancer" includes, squamous cell . carcinoma, adenocarcinoma and undifferentiated non-small cell lung cancer (undeveloped cancer cells are known as undifferentiated cells) and large cell carcinoma.
"Colorectal cancer" as used herein, includes any type of colon or rectal cancer, including but not limited to, adenoscarcinoma, sarcoma, melanoma, stromal, carcinoid, and lymphoma. '
In one embodiment, the present invention is a method of treating a patient suffering from an acute myeloid leukemia characterized by a FLT3 mutation.
In certain embodiments, the compound of formula (I) can be administered in combination with an anti-cancer agent.
The compounds used in the present invention can be administered alone or in combination with one or more other pharmaceutically active agents that are effective against the cancer being treated.
As used herein, the term "combination" with reference to pharmaceutically active agents and the term "co-administering" and "co-administration" refer to administering more than one pharmaceutically active agent to a patient during one treatment cycle and not necessarily simultaneous or in a mixture.
Anti -cancer agents that can be employed in combination with the compounds of the invention include Taxol™ (also referred to as "paclitaxel", and compounds that have the basic taxane skeleton), Adriamycin, Dactinomycin, Bleomycin, Vinblastine, Cisplatin, acivicin; aclaπibicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; 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; cedefmgol; chlorambucil; cirolemycin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; doxorubicin; doxorubicin 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; fiudarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin II (including recombinant interleukin II, or rIL2)3 interferon alfa-2a; interferon alfa-2b; interferon alfa-nl ; interferon alfa-n3; interferon beta-I a; interferon gamma-I b; iproplatin; 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; mitocromiπ; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; piicamycin; plomestane; porfimer sodium; porfiromycin; prednimustinε; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safmgol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatjn; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; 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 that can be employed in combination with the compounds described herein include: 20-epi-l,25 dihydroxyvitamin D3; 5- ethynylurac.il; abiraterone; aclarύbicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; ainidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; ' andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti- dorsalizing morphogenetic 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-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine; carboxamide-amino- triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprostj.cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; colIismycin 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; 9- dioxamycin; diphenyl spiromustine; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; efiornithine; 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; fotemustiπe; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilαmastat; imidazoacridones; 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; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; xnerbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin analogues; mitonafϊde; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotropin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1 -based therapy; 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; neutral endopeptidase; nihitamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; O6-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral' cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; 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; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum compounds; platinurή-triamine complex; porfϊmer 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; RII retinamide; rogletimide; 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; signal transduction modulators; single chain antigen-binding protein; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1 ; squalamine; stem cell inhibitor; stem-cell division inhibitors;- stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide: tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin- receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; 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; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer. Preferred anti-cancer drugs are 5-fluorouracil-and leucovorin.
Other xhemotherapeutic agents that can be employed in combination with the compounds of the invention include but are not limited to alkylating agents, antimetabolites, natural products, or hormones.
In the embodiments, in which the compounds of the invention are used to treat cancer, the dosage range at which the disclosed compounds, for example, compounds- of formula (I)5 including the above-mentioned examples thereof, exhibit their ability to act therapeutically can vary depending upon the severity of the condition, the patient, the formulation, other underlying disease states that the patient is suffering from, and other medications that may be concurrently administered to the patient. Generally, the compounds described herein will exhibit their therapeutic activities at dosages of between about 0.1 mg/m2 free base equivalent per square meter of body surface area/single dose to about 1000 mg/m2 free base equivalent per square meter of body surface area/single dose. For example, the single dosage range can be between 10-800 mg/m2, 100-700 mg/m2, 400-600 mg/m2, 420-550 mg/m2 or 440-500 mg/m2 or the single dose can be 480 mg/m2.
In treating a patient afflicted with a conditions described above, all of the disclosed compounds can be administered in any form or mode which makes the compound bioavailable in therapeutically effective amounts. For example, compounds of formula (I) can be administered in a form of a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salts" means either an acid addition salt or a basic addition salt, whichever is possible to make with the compounds of the present invention. "Pharmaceutically acceptable acid addition salt" is any non-toxic organic or inorganic acid addition salt of the base compounds represented by formula (I). Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids which form suitable salts include the mono-, di- and tricarboxylic acids. Illustrative of such acids are, for example, acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, salicyclic, 2- phenoxybenzoic, p-toluenesulfonic acid and sulfonic acids such as methanesulfonic acid and 2-hydroxyethanesulfonic acid. Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated or substantially anhydrous form. In general, the. acid addition salts of. these compounds are more soluble in water and various hydrophilic organic solvents and which in comparison to their free base forms, generally demonstrate higher melting points. "Pharmaceutically acceptable basic addition salts" means non-toxic organic or inorganic basic addition salts of the compounds of formula (I). Examples are alkali metal or alkaline-earth metal hydroxides such as sodium, potassium, calcium, magnesium or barium hydroxides; ammonia, and aliphatic, alicyclic, or aromatic organic amines such as methylamine, trimethylamine and picoline. The selection of the appropriate salt may be important so that the ester is not hydrolyzed. The selection criteria for the appropriate salt will be known to one skilled in the art.
Compounds of the present invention can be administered by a number of routes including orally, sublingually, buccally, subcutaneously, intramuscularly, intravenously, transdermally, intranasally, rectally, topically, and the like. One skilled in the art of preparing formulations can determine the proper form and mode of administration depending upon the particular characteristics of the compound selected for the condition or disease to be treated, the stage of the disease, the condition of the patient and other relevant circumstances. For example, see Remington's Pharmaceutical Sciences, 38th Edition, Mack Publishing Co. (1990), incorporated herein by reference.
The compound of formula (I) of this invention may also be administered topically, and when done so the carrier may suitably comprise a solution, ointment or gel base. The base, for example, may comprise one or more of petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil,- diluents such as water and alcohol, and emulsifϊers and stabilizers.
The solutions or suspensions may also include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylene diaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampules, disposable syringes or multiple dose vials.
The invention is illustrated by the following examples, which are not intended to be limiting in any way.
EXEMPLIFICATION
Example 1 Svmadex™ inhibits proliferation ofB-Cells following stimulation with LPS and T-CeIIs following stimulation with Con A in in vitro experiments
The activity of Symadex™ was compared to mitoxantrαne in several in vitro assays to determine the effect of Symadex™ on several key regulatory systems involved in multiple sclerosis neuroinflammation and antigen presentation.
IL-4 serves as a growth and differentiation factor for B cells, mast cells and macrophages and is a switch factor for synthesis of IgE in mice. It also promotes growth of a cloned CD4+ T cell and enhances class II MHC molecule expression and resting B lymphocytes enlargement. In man, CD4+ T lymphocytes also produce IL- 4, but the human variety has not been shown to serve as a B cell or mast cell growth factor. Both murine and human IL-4 induce switching of B lymphocytes to synthesize IgE. Human IL-4 also induces CD23 expression by B lymphocytes and macrophages in man. IL-4 may have some role in cell mediated immunity.
IL-IO inhibits cytokine synthesis by THI cells, blocks antigen presentation, and inhibits the formation of interferon γ. IL-10 inhibits the macophage's ability to present antigen and to form IL-I, IL-6 and TNF-α. IL-10 also participates in IgE regulation. Although IL-10 suppresses cell-mediated immunity, it stimulates B lymphocytes, IL-2 and IL-4 T lymphocyte responsiveness in vitro, and murine mast cells exposed to IL-3 and IL-4. IL-10 may find therapeutic utility by suppressing T lymphocyte autoimmunity in multiple sclerosis and type I diabetes mellitus as well as in facilitating allograft survival.
In this experiment, test compound and/or vehicle were preincubated with human peripheral blood mononuclear leukocyte (PBML, 1 x 1O6AnI) in RPMI buffer pH 7.4 for 2 hours. Concanavalin A (Con A, 20 μg/ml) was then added to stimulate the cells overnight in 5% CO2 at 370C. IL-4 and IL-IO cytokine levels in the conditioned medium were then quantified using a sandwich ELISA kit. Compounds •were screened at 10, 1, 0.1, 0.0,1 and 0.001 μM.
B-lymphocyte cells isolated from the spleen of balb/c mice weighing 17 + 1 g were used. Test compound and/or vehicle were incubated with the cells (1.5 x 106/ml) in the presence of 10 μg/ml lipopoly saccharide (LPS) in AIM-V medium pH 7.4 at 370C for 24 hours. [3H3Thymidine (120 nM) was then added for an additional overnight incubation period. Thymidine incorporation was assessed by liquid scintillation counting.
T-lymphocyte cells isolated from thymus of balb/c mice weighing 17 ± 1 g were used. Test compound and/or vehicle is incubated with the cells (4 x 106/ml) in the presence of 3 μg/mL Concanavalin A (Con A) in AIM-V medium pH 7.4 at 370C for 24 hours. [3H]Thyrmdine (120 nM) was then added for an additional overnight incubation period. Thymidine incorporation was assessed by liquid scintillation counting.
Compounds were screened at 10, 1 , 0.1, 0.01 and 0.001 μM.
The results for mitoxantrone and Symadex™ are presented in Table 1. Test compound-induced suppression of cell proliferation by 50 percent or more (≥50%) relative to vehicle control response indicates significant inhibitory activity.
Figure imgf000044_0001
Figure imgf000045_0001
The results presented in Table 1, FIG. IA and IB as well as in FIG. 2A and 2B indicate that Symadex™ inhibits the release of inflammatory mediators IL-4 and IL-10, both of high importance in neuroinflammatory diseases such as multiple sclerosis. Furthermore, Symadex™ exhibited high level of growth inhibition in in vitro proliferation, assays involving B- and T-cells. The activity of Symadex™ was comparable to that of the. control compound, mitoxantrone. Example 2 Symadex™ Alleviates the Symptoms of .Experimental Autoimmune Encephalomyelitis (EAE), an Animal Model of chronic Multiple Sclerosis in a 'weekly treatment cycle for 4 weeks
" One method of showing the utility of the a pharmaceutical compound for the treatment of various conditions associated with multiple sclerosis (MS) is its ability .to inhibit effects of Experimental Autoimmune Encephalomyelitis in laboratory animals.
Experimental Autoimmune Encephalomyelitis (EAE) is an animal model for MSS which entails inducing a T-cell-mediated autoimmune disease against myelin basic protein in certain susceptible mammalian species. The EAE model is an appropriate method for studying the inflammation of the brain and spinal cord associated with MS (see Bolton, C. Mult, Scler, 1995; 1(3); 143 -9).'
In rodents, injection of whole spinal cord or spinal cord components such as myelin basic protein induces an autoimmune response based on the activation of T- lymphocytes. Clinical disease typically becomes manifest around day 8-10 after inoculation, observed as a broad spectrum of behavioral anomalies ranging from mild gait disturbances and tail atony to complete paralysis and death. Weight loss typically occurs. In animals that survive, spontaneous recovery occurs, accompanied by variable recovery of most motor function. Depending on the species, allergen, and methodology used, animals tested by the EAE model may experience a single (acute EAE) or several (chronic relapsing EAE) attacks. ■
Treatments of EAE come in many structural forms: treatment can be prophylactic or preventative, whereby the therapeutic composition is administered before immunization; treatment can be initiated during the first week of induction; and treatment can be interventious, initiated after clinical symptoms are extent (acute or chronic). Prevention protocols are very common in the literature, treatment after disease is rarer, and treatment after weeks of disease are the most infrequent. The experiments reported herein are in the last classification in which animals in the chronic- progressive (CP) phase with extensive demyelinated plaques are treated. CP- EAE induced by whole CNS in complete Freund's adjuvant is a florid disease with extensive inflammatory and demyelinated changes. As a general philosophy, we believe that successful intervention at later times can better predict effectiveness in the human condition. This is particular relevant to the case of prevention studies, which concentrates on the peripheral immune system, rather than addressing the issue of existing CNS. inflammation that is a characteristic of MS.
Methodology
In the present experiment, female juvenile Hartley guinea pigs (225 g) were immunized with homogenized whole CNS (in saline) with an equal amount of complete Freund's Adjuvant and 10 mg added killed M, tuberculosis. The animals (>95%) show clinical signs starting on day 7 post immunization. An acute eveniof varying severity occurs between day 7 and day 20 followed by a continuous accumulation of clinical abnormality with hind limb paralysis, fecal impaction and incontinence. Table 2 shows the clinical scoring scale. These clinical features indicate inflammation — induced lumbar spinal cord demyelination. A recent survey of previous experiments indicates that taking an animal past day 40, which has a clinical score of "2" for more than 1 week yields a 97% occurrence of demyelinated plaques in the cord.
In these experiments, the immunized animals were nursed until day 40 or day 52 and then treated with 8 mg/kg and 16 mg/kg Symadex™ (intra cardiac), or 20 mg/kg and 40 mg/kg Symadex™ (i.p.) once a week for 4 weeks. Controls were given vehicle. Clinical signs were scored daily and the weights recorded. At the completion of the treatment period, the brain and spinal cord were dissected, formalin fixed and blocked for routine pathological examination of meningeal inflammation, perivascular infiltration (cuffing), parenchymal myelitis and demyelination by a blinded observer using hematoxylin-eosin and solochrome R cyanin stained sections.
Untreated, chronic EAE animals (n=5) were sacrificed on day 40, as well as non-E AE controls (n=5). Following each 10-day treatment interval, five animals from each group were sacrificed (0.25 ml sodium pentobarbital), blood samples were collected for FACS analysis (see below), and the brain and spinal cord dissected and sectioned. Three spinal sections were used, corresponding to lumbar, thoracic and cervical regions of the cord. The brain was cut into five transverse sections; the first three proximal sections .were combined in one block, and the last two distal sections in another. Tissues were fixed in 10% formalin and embedded in paraffin. Five micrometer sections were stained with hematoxylin-eosin (H-E) or solochrome-R-cyanin (SCR) and evaluated by a blinded observer in each of the four categories: meningeal inflammation, perivascular infiltration, encephalitis or myelitis and demyelination (Table 2). The combined pathological score represents the total score (out of a potential 20) from all five CNS sections in each animal.
Table 2
Pathological scoring scale
M: Inflammatory reaction in the meninges
0: no changes
1 : perivascular and/or meningeal infiltration by mononuclear cells, 1-3 vessels involved 2: 4-6 vessels involved
3: 6+ vessels involved
4: dense infiltration of meninges with nearly all or ail blood vessels involved
P: Parenchymal perivascular infiltrations
0: no changes
1 : 1 -3.parenchymal vessels infiltrated in Virchow-Robin spaces
2: 4-6 vessels involved
3 : 6+ vessels involved
4: virtually all vessels involved
E: Encephalitis or myelitis
0: no invasion of the neural parenchyma; microglial or inflammatory cells invading neural parenchyma 1 : a few scattered cells
2: invasion by cells from several perivascular cuffs
3: large areas of neural parenchyma involved
4: virtually the entire section is infiltrated
D: Demyelination, remyelination and myelin debris
0: no demyelination
1 : single focus on subpial demylination or myelin debris
2: several small foci of demyelination
3: one large confluent area of demyelination
4: several large confluent areas of demyelination _______ To quantify the abnormalities observed in the spinal cord, sections stained with H-E were divided into 12 representative pie-shaped areas. In each area, the number of cells within a 0.12-mm2 field of view was counted using Sigma Scan Pro image analysis software (SPSS), and the combined mean number of cell in all 12 areas was calculated for the whole spinal cord (36 fields of view per- animal). Note that as all cell nuclei were counted, the number of cells may include neurons and glial cells in addition to infiltrates. Hence, the cell count in non-EAE animals served as a baseline.
Results
Symadex™ produced a profound and substantial change in the clinical progress and pathological findings when given at 20 and 40 mg/kg (i.p.). FIG. 3 illustrates the mean clinical score of the animals at the indicated day thereafter. The treated animals all showed some degree of clinical recovery with the 40 mg/kg group reaching recovery within 2 weeks of the start of treatment.
The longitudinal course of recovery from disease is further illustrated in FIG." 4. In this experiment, the vehicle controls showed a steady course of disease. Treated animals, in both the 20 and 40 mg/kg cohorts entered the study presenting disease of greater severity than controls, a coincidental circumstance attributed to the phenomenology of randomization prior to assignment of a treating group. As indicated by the arrows, treatment began on day forty and progressed for a total of four doses. At the end of the treatment, both treated cohorts showed significant disease improvement to levels significantly below control, despite having entered the study at a disease level well above control. Statistical significance, even with 3 and 4 animal small cohorts showed p values supporting the hypothesis that treatment afforded a statistically different outcome over control. These were determined by non-parametric comparisons by the Mann- Whitney or Wilcoxon rank-sum test for difference in medians. The significance values were 0.001 and 0.004 for the 40 and 20 mg/kg cohorts, respectively.
The pathological findings were most unusual. The scores for meningeal ' inflammation and perivascular infiltration were more severe in the treated groups than in vehicle controls (data not shown). However, we observed two highly significant findings: existing lesions had a profound loss of cells (data not shown) and we observed myelin pallor previously attributed to remyelination (data not shown). The latter observation is consistent with permissive remyelination of the CNS due to removal of the inflammatory cells.
Demyelination is a key pathological feature of the MS lesion..Not only does this alter electrical response of the axon, current thought suggests that prolonged demyelination can result in permanent axonal damage and death. Thus neuro- degeneration is also a key component of the MS pathological milieu. In this regard, Symadex™ has proved to be effective in permitting endogenous remyelination even after a period of disease progression that reached 97% spinal chord demyelination in this chronic - progressive model. It appears to permit this CNS recovery by reducing the inflammation in existing lesions. After prolonged Symadex™ treatment, it-is possible to observe chronic demyelinated plaques that have virtually no' remaining inflammatory cells and some of these lesions show the myelin pallor indicative of remyelination (called a shadow plaque in MS). Prevention of new T- cell infiltration by deletion of these cells or down regulation or inhibiting cell trafficking would prevent the recruitment of further macrophages to an inflammatory lesion. As a consequence, the immune cells in the lesions die by apoptosis and the lesions are left relatively free of infiltrates. Removal of the cytokine, and ROS — mediated tissue toxicity of macrophages would allow the CNS reparative mechanisms to become active and remyelination is observed. It is thus likely that Symadex™ has an effect on the peripheral immune system, although a direct effect on CNS inflammation cannot, be ruled out.
The continued presence of large inflammatory cuffs and meningeal inflammation scores that were higher than control is consistent with the continued production of immune competent leukocytes which accumulate around CNS vessels, but do not traffic into the parenchyma. Example 3 Symadex™ Alleviates the Symptoms of Experimental Autoimmune Encephalomyelitis (EAE), an Animal Model of chronic Multiple Sclerosis in a weekly treatment cycle for 4, 6. 8 and in a 4 week on drug-4 week off treatment cycle
The experiment described in Example 2 was extended to a larger cohort and longer treatment cycle with several objectives in mind. In addition to corroborating the initial findings, a concerted effort was directed at also demonstrating the extent and durability of response, including the effect after drug withdrawal, and to document the impact of drug treatment on immune function in order to uncover any signals of impending impairment oτ toxicity.
Following disease induction, as previously described, the animals were randomized into 5 five cohorts, one vehicle control and 4 treatment cohorts. Animals in the treatment cohorts were administered study drug intraperitoneally at 20 mg/kg (Symadex™dihydrochloride trihydrate) once a week for 4, 6 and 8 weeks, with an additional cohort treated once a week for 4 weeks and observed for an additional 4 weeks of treatment with vehicle solution (saline) rather than with drug.
The only significant protocol deviation from the method of Example 2 was that the pool of immunized animals was culled of animals presenting with a' disease severity greater than 2 and randomized so that the' mean disease severity of each cohort was matched in the severity score range of 1 to 1.5. This measure was invoked in order to avoid the chance circumstance, observed in Example 2, that animal selected for treatment should start treatment with a more severe presentation than the corresponding vehicle controls.
AU treated animals showed statistically significant improvement in disease. That is, their symptoms of paralysis attributable to the demyelinating progression of inflammatory cell assault on nerve chord parenchyma, were reduced close to baseline, pre-disease levels. These results are evident by mere inspection of the disease course plots and also proved to be highly significant by non-parametric, rank-order statistical analysis.
The 4-week treatment cycle result (n=l 4), as shown in FIG 5, demonstrates a declining trend in clinical severity score to a mean level 0.7 from a starting disease presentation mean of 1.3. This change is statistically different from control Ov=I 3), with a p value on differences in median of 0.0001. A similar dose response is demonstrated in FIG 6, which describes the 6 week treatment course. Again, disease improved from a mean severity score of 1.3 for control (π=4) down to 0.3 (n=3) for •the treatment cohort, with a statistical significance. p value of 0.009. FIG. 7 presents the 8 week treatment cohorts. Attention is called to the vehicle control, whose disease shows progression towards greater severity, and.hence greater paralysis attributable to demyelination, with a gradual rise after 4 weeks from a level of 1.3 to 1.7. In contrast, both treatment cohorts, show progressive disease amelioration, indication reversal of demyelination. In the case of animals treated with 8 consecutive doses, the trend towards normal;, pre-disease clinical scores is initially variable but converges on baseline at the end of the treatment course. Despite the interindividual heterogeneity in recovery profile, the full rank-order analysis shows the difference between treatment (n=3) and control (n=5) to be significantly at a p value of 0.0006. The cohort with treatment discontinued treatment after 4 weeks, remained in stable disease at the time of discontinuation, also at a level significantly different from control, with a p value of 0.002.
The results of this study are shown in FIG. 7. First, the therapeutic response shows a graded temporal response against control. Sick animals become progressively healthier in proportion to the weekly duration of response, while . control animals progress irreversibly towards complete neurological dysfunction, whose root cause is irreversible demyelination. Second, the cumulative pharmacodynamic effect of drug treatment is durable, because treated animals remain in stable condition, commensurate with their degree of treatment, while untreated controls sustain an accelerating progression in. disease. This is a particularly noteworthy observation in light of the effects obtained with the' most promising recent therapy for progressive MS, namely the α4 integrin antagonists like Tysabri and its small molecule ligand equivalents, as described by Piraino PiS. et al, J. Neuroimmunology, 131: 147-159, 2002). When treatment is discontinued in the same guinea pig EAE model of MS, the animals in the treatment cohort revert within 7 days to the disease level of untreated controls, with no evidence of a protracted beneficial pharmacodynamic effect.
. The contrasting result between the therapeutic effects of Symadex and the α4 integrin antagonists, as well as between Symadex and other therapies that interdict T-cell mediated inflammatory responses, is that Symadex does not exert its action via the activation and recruitment of inflammatory cells. The histopathology of spinal chord from animals sacrificed at periodic interval throughout the time course of disease recovery show accumulation, rather than diminution, of inflammatory cells in blood vessels and perivascular cuffs, as noted in Example 2. Yet, these cells are apparently blocked from transmigrating beyond the basement membrane of parenchyma, suggesting a block via mechanisms that could involve: cell adhesion, motility, and extracellular matrix remodeling.
It is demonstrable as a differentiating, and unexpected, feature of Symadex' s mode of action, when compared to corticosteroid, interferon, and integrin antagonist therapies that there are no changes in T-cell populations or in T-cell sub-type ratios.' In the instant example, as shown in FIG. 8, Panel A5 no difference is observed between any treatment cohort and vehicle control with respect to total T-cell counts. Neither are CD4 or GD8 T-cell populations modulated by Symadex™ treatment, as shown in panels B and C. Even B-cells show no significant deviation from controls, although there appears to be a declining trend across' the board as a function of disease or treatment duration. Like the cytotoxic therapies for MS, e.g. mitoxantrone and cyclophosphamide, for example, Symadex™ may exert a transient diminution in B-cell counts in stimulated cell cultures, according to the evidence presented in Example 1. but the effect is not evident on prolonged exposure in vivo. This observation further reinforces the notion that Symadex™ acts by a novel mechanism that will not deplete the immune system nor diminish host defenses against antigen presenting pathogens. Such a propeπy would be highly desirable and advantageous in any therapy intended for the treatment of chronic conditions or acute conditions, such as MS flair ups, in otherwise healthy subjects.
Example 4 Svmadex™ Reverses the Symptoms of Experimental Autoimmune Encephalomyelitis (EAE), an Animal Model of chronic Multiple Sclerosis after two doses administered 72 hours apart. ' '
Analysis of the time course of disease recovery upon treatment with Symadex™ on a weekly basis reveals a two to three day periodicity in the • amelioration of disease. This phenomenon is particularly evident in the 8 week treatment test cohort shown in FIG 7. Individual animals in the cohort can be seen to respond differently so that the pharmacodynamic response on average presents itself in a "saw-tooth" pattern between successive dosing intervals. In tracing the records of specific animals within the cohort, it appears that some recover .transiently and show disease improvement for 2-3 days after receiving a dose and then revert to a higher disease score. The overall trend leads to disease improvement over 8 weeks, but the "saw-tooth" response phenomenon raises questions about the temporal spacing of treatments that optimally achieve a smoother reversal of disease symptoms.
In order to test the possibility that- a more frequent dosing schedule would offer a more rapid resolution of disease symptoms, an experiment was performed to match the dosing cycle to the observed periodicity of response. Accordingly, the method of Experiment 2 was applied to a cohort of animals and controls, which were allowed to reach' the chronic phase of disease at.30 days post immunization. Six animals with a disease score of 1 were selected and half were treated -with 20 mg/kg Symadex™ administered intraperitoneally. Two dose were given 72 hours apart to 3 animals. Three animals served as vehicle controls.
As shown in FIG. 9, the Symadex™ treatment on a 72 hour schedule reversed the progression of disease and restored baseline clinical scores with just 2 doses, while disease progression continued in the control cohort. The difference is statistically significant by a p value of 0.002 by the rank-sum test.
" This experiment demonstrates that a more frequent dosing of Symadex™ can be tailored to match the particular balance between drug residence time, the temporal properties of the assault by inflammatory cells on myelin, and the intrinsic processes of permissive remyelination. It would be reasonable, therefore, to expect that combinations of dosing regimens can be applied first to accelerate recovery from disease, by more frequent or intense schedules of drug delivery, and then to maintain the beneficial effects of inflammatory cell blockade with less frequent, but, periodic, booster doses. The "saw-tooth" patterns of treatment and disease reversion evidenced in FIG. 7, in Example 3, suggested that the effect of Symadex™ can be attenuated over a 2-3 day interval between doses. This experiment confirms that the efficacy of Symadex™ can be re-enforced through more frequent dose administration.
Example 5 Symadex™ Alleviates the Symptoms of Experimental Autoimmune Encephalomyelitis (EAE), an Animal Model of Acute Multiple Sclerosis Upon Daily Dosing Over the Initial Course of Disease Induction.
As described in Example 2, the EAE model in the guinea pig is biphasic. After the myelin basic protein insult on initial immunization, the typical clinical pattern of neurological impairment begins with acute signs of disease day 9 post immunization. Clinical onset results in weight loss, hind limb weakness and an abnormal righting reflex. The severity of these symptoms peaks over 6-7 additional days followed by a short duration transient and partial resolution by day until day- 20, when the disease course changes to a steady progressive decline, from which there is no clinical recovery.
As an important extension to the utility of Symadex, its therapeutic effect at this earlier stage of disease presentation was examined. The experiment was further designed to build on the results of Example 4, which suggested that more frequent dosing affords more rapid and unidirectional symptom resolution. Since the acute phase of EAE also mimics active, but not necessarily progressive disease, as would be expected to be the case in human subjects with remitting-relapsing multiple sclerosis, the experiment was further designed to test the efficacy in comparison to mitoxantrone. This later drug, as indicated earlier, is an approved therapeutic agent and had served as the starting point for the chemical evolution of what became the Symadex™ molecule minus the toxicophoies known to be causative agents for cardiotoxicity.
Accordingly, three randomized cohorts of guinea pigs with EAE induced by the method of Example 2, were treated, respectively, with 6 mg/kg of Symadex™ (full salt hydrate) and 0.35 mg/kg of mitoxantrone. Animals were treated daily, by intraperitoneal injection, for 15 days, starting on day 7 post immunization. Controls were treated with vehicle. We reasoned that 15 consecutive, 6 mg/kg doses of Symadex™ would represent a level of drug exposure that would be commensurate with the "20 mg/kg every 72 hours" regimen in Example 4 and consistent with a mid level exposure between the 20 mg/kg and the40 mg/kg schedule given weekly, in Example 2. The mitoxantrone .dose was selected to reflect a typical high dose given to rats or mice by daily dosing in the prior art, but allonietrically scaled to the guinea pig- As can be appreciated from the results presented in FIG. 10, the cohorts treated with either Symadex™ or mitoxantrone present a consistently different trend than the vehicle controls. In the controls, the onset of disease is followed by a steady increase in clinical score until day 15, followed by the characteristic short term reversion and a second climb towards higher disease severity by day 20. In the case of both mitoxantrone and Symadex, the initial rise in neurological impairment is arrested, and all animals continued on a course of recovery towards basal levels throughout the dosing period. However, statistical analysis by Mann- Whitney rank- sum tests indicates that the therapeutic effect, of Symadex™ against both control and the performance of mitoxantrone reaches statistical significance with a p value below 0.05. The difference in median scores between the performance of mitoxantrone and control did not reach statistical significance. FIG. 11 shows the weight gain profile, a sensitive indicator of general health status in guinea pigs. Acute EAE disease onset triggers a rapid weight loss from which there develops a steady recovery after day-15. Control animals and Symadex™ treated animals regain the ability to add weight every day, which is the norm for guinea pigs when healthy and prior to the onset of severe, chronic disease. However, under the circumstances of this experiment, mitoxantrone may have alleviated the acute clinical symptoms of EAE at a lower relative dose but it also impairs weight gain, a sign of generalized toxicological response to a drug that is a potent and broad spectrum cytotoxic. Comparison of the weight gain profiles between vehicle controls and Symadex™ did not show statistical significance by the Mann- Whitney rank-sum test, whereas the difference between Symadex™ and mitoxantrone reached statistical significance with a p value of 0.034. .
These results confirm that Symadex™ modifies the presentation of EAE throughout the course of active disease, both at the early acute and at the chronic phase without imposing a deleterious cytotoxic load. An analysis of the pathophysiology, shown in FIG 12, confirms the earlier observations, by the histological methods described in Example 2, that Symadex™ arrests the invasion of parenchyma by inflammatory cells. It does so with a significant difference from the mode of action of mitoxantrone and drugs in the mitoxantrone class which are immunosuppressive. While the outcome may be the same in terms of the effect of both drugs on reducing the perivascular cuffing (P), myelitis (E) and demyelination (D), as judged by statistically significant differences (p value <; 0.05) from control, Symadex™ and mitoxantrone do not show the same action on meningeal inflammation (M). Mitoxantrone is an immunosuppressant that blocks activation and recruitment of inflammatory cells given the statistically significant reduction in meningeal inflammation (M). Symadex™ does not, although the therapeutic outcome according to the remaining three histopathological assessments is essentially similar.
These findings are relevant to the human disease circumstance, because it is considered highly beneficial to effect treatment of MS conditions without impairing the host's ability to mount an immunological, and hence inflammatory response, against adventitious infections. It terms of response to cytotoxic agents, which might impair gastrointestinal function and nutritional maintenance, the lack of negative effects on normal growth and weight gain in these guinea pig experiments points to another safety advantage that may accrue to Symadex™ therapy. The cumulative 15 day dose of Symadex™ for treatment of acute, active disease is 90 mg/kg. Allometric scaling to human dosimetry levels yields a corresponding human dose of 540 mg/m2 body surface area (as free base), which has been shown to be a safe and well-tolerated single dose, and is lower τhan the 640 mg/m2 dose which is indicated as a repeat dose every three weeks. Allometric scaling of the mitoxantrone dose, on the other hand, represents a total human equivalent dose of 45 mg/m2. Since mitoxantrone is used in the treatment of MS on a three month dosing cycle at 12 mg/m2, this represents the total dose for a year's worth of treatment. Thus, the experimental findings in this comparative example on the relative efficacy of Symadex™ versus mitoxantrone suggest that in humans a single dose of Symadex™ should show a similar, if not greater, therapeutic benefit as an year's course of mitoxantrone. Example 6 Svmadex™ Alleviates the Symptoms of Collagen Antibody Induced Arthritis in the Mouse, an Animal Model of Rheumatoid Arthritis and Autoimmune Disease
Rheumatoid Arthritis (RA) is an autoimmune disorder characterized by the chronic erosive inflammation in joints leading to the destruction of cartilage and bones. Several disease modifying antirheumatic drugs (DMARDS) are used in the treatment of RA. Currently, the two most important DMARDS are inhibitors of tumor necrosis factor α (TNF-α) and methotrexate (MTX). One method for demonstrating the utility of a pharmaceutical compound for the treatment of various conditions associated with RA is its ability to inhibit the induction of arthritis by collagen monoclonal antibodies (mABs) in mice.
Collagen-induced Arthritis (CIA) is an experimental autoimmune disease that can be elicited in susceptible strains of rodents (rat and mouse) and nonhuman primates by immunization with type II collagen, the major constituent protein of articular cartilage. CIA manifests as swelling and erythema in the limbs of the mouse. This model of autoimmunity shares several clinical and pathological features with rheumatoid arthritis (RA) and has become the most widely studied model of RA. CIA in the mouse model was first described by Courtenay et al. in 1980 (Courtnay, J.S., Dallman, MJ., Dayman, A.D., Martin A.s and Mosedale, B. (1980) Immunisation against heterologous type II collagen induces arthritis in mice. Nature 283, 666-668). Like RA5 susceptibility to CIA is regulated by the class II molecules of the major histocompatibility complex (MHC), indicating the crucial role played by T cells.
Methods
Groups of 3 BALB/c strain mice, 6-7 weeks of age, were used for the induction of arthritis by monoclonal antibodies (mABs) raised against type II collagen, plus Iipopolysaccharide (LPS). A combination of 4 different mABs (D8, Fl O, DI-2G and A2) totaling 4 mg/mouse was administered to the animal . intravenously on day 0, followed by intravenous challenge with 25 mg/mouse of LPS 72 hours later (day 3). From day 3, test substance and vehicle were each administered orally once daily for 3 consecutive days. For each animal, volumes of both hind paws were measured using a plethysmometer with water cell (12 mm diameter) on Days 0, 5, 7, 10, 14 and 17. Percent inhibition of increase in volume induced by mABs + LPS was calculated by the following formula:
Inhibition (%): [1 - (Tn - T0)/(Cn - CO)] x 100%
Where: " " CO (Cn): volume of day 0 (day n) in vehicle control
TO (Tn)* volume of day 0 (day n) in test compound-treated group
Reduction of edema in the hind paws by 30% or more is considered significant.
'Results
To monitor the onset of CIA, the volume of the two hind paws of mAB treated mice were measured. In the control (vehicle) treated animals the paws quickly became inflamed with a 42% increase in volume on day 5, the maximum volume was observed on day 10 and then theN swelling began to subside. As shown in FIG. 13, in . the Symadex™ treated group, the initial swelling on day 5 was slightly lower then control (32% vs. 42%) and significantly less inflammation (measured by paw volume) was observed on day 10 (29% vs. 75%), day 14 (18% vs. 70%) and day 17 (19% vs. 47%). The difference in means by paired t-test and in medians by non- parametric Mann-Whitney rank-sums all show p values lower than 0.01.
Conclusion
Symadex™ demonstrated significant anti-arthritic activity in the mouse CIA model, with significant anti-inflammatory activity on day 10 (61% inhibition), day 14 (74% inhibition) and day 17 (59% inhibition). These findings are relevant in the context of prior example on EAE and autoimmune disease in general because they exemplify the efficacy of Symadex™ via an unexpected mechanism. The collagen antibody model of rheumatoid arthritis is significant because it by-passes the primary inflammatory insult of antigen presentation. Classical anti-inflammatories like the corticosteroids and anti -folates like methotrexate, alleviate the consequence of autoimmune inflammatory diseases by suppressing the primary events of inflammatory cell activation and recruitment. The antibody induced model generates the symptoms of disease that present in the later stages of the autoimmune response, after activated cells become invasive into cartilage, having extravasated and transmigrated, as would be the case in MS during a prolonged assault on parenchyma.
Methotrexate, a benchmark therapeutic agent, has been shown to yield diminishing benefit in antibody induced models, which are intrinsically less dependent on T-cell activation than on their trafficking and migratory properties. The work of Lange et al. can be cited in this context {Annals of Rheumatoid Disease 64:599-605, 2005). By contrast, Symadex™ appears fully active in this model. The results presented in this example are especially relevant to the treatment of human subjects, because the therapeutic effect was obtained by oral administration. In the era of injectable biologies, such as blocking antibodies, the addition of an effective, non-immunosuppressive therapy via the oral route is particularly desirable.
Example 7 Symadex"™ downresulates otherwise overexpressed target mechanisms of inflammatory eel! adhesion, cell-surface signaling, and cell proliferation
To explore the effect of Symadex™ treatment on gene expression, microarray experiments were performed.
Two colorectal cancer cell lines (HT29 & HCTl 16) were chosen for study, whose behavior as rapidly proliferating invasive cells could be generalized to many other such cell types from different tissue origins. The two lines were immortalized colon carcinomas. Their gene expression patterns are known to mimic the behavior of n euro-enteric cells and therefore provide an appropriate simulation of the kinds of regulatory patterns that would be found in cells of similar epithelial or endothelial origin. Cells with these ontological roots are also suitable models for the kinds of autoimmune and inflammatory susceptibilities that are common in tissues of neuroenteric origin, such as those in which inflammatory bowel disease woμld present itself.
Attention is drawn here to the exhaustive studies, using differential gene expression arrays (Zhang J. et al., "Neural system-enriched expression: relationship to biological pathways and neurological diseases", Physiol. Genomics 18:167-183, 2004) which have documented the redundancies and commonalities of gene expression .patterns in both the central nervous system and in anatomically unrelated tissues. For example, Zhang and colleagues, whose teachings are incorporated here by reference, profiled the expression products of 8,734 genes in 10 regions of the nervous system and in 30 peripheral organs. Their analyses reveal that approximately 70% of the genes relevant to nervous system diseases are also expressed in multiple tissues, including those of epithelial origin and in peripheral blood. These "investigators suggest further that the profiling of genes implicated in nervous system diseases but sourced from various peripheral tissues, where easier sampling can be obtained, will aid the development of better mechanistic understanding about those diseases. Hence, the use of colon cells in gene expression studies as a model paradigm for understanding the effect of a drug on pathways . common to those cells and nervous system tissues is experimentally justifiable.
Accordingly, the specific studies to document the mechanism of action of the compounds in the instant invention, were conducted as follows using the preferred imidazoacriήidone composition, referred to hereinafter as Symadex™.
Cells were grown in the presence of Syrnadex™ at the GI50 concentration (0.68 and 0.21 μMolar, for the HT29 and HCTl 16 cell lines, respectively), and harvested along with untreated control fractions after 1, 8 and 48 hrs. of exposure. Frozen cell pellets were lysed'in triplicate and total R]MA isolated by purification over spin columns (all reagents from Ambion). After QC acceptance for purity, total RNA was converted to cRNA by linear amplification and 10 μg samples were • applied to CodeLink Human Whole Genome Bioarrays (GE Healthcare and'GenUs Biosystems).
Arrays were processed in triplicate and comparisons made after robust statistical analysis of replicate variability. Genes (including ESTs) were considered to be differentially expressed if a change from baseline could be demonstrated as .significant by T-test (p<0.05' 5 <x=0.025), using CodeLink Expression Analysis (GE Healthcare) and GeneSpring (Silicon Genetics) software. False discovery rates and representation in standardized gene ontologies/pathways were then determined by filtering the "fold" changes, in expression with open access software packages, EASE and GoMiner, and with Pathways Analysis (Ingenuity Systems). Functional annotations were then explored further in the MedMiner, literature search environment.
Over the interval sampled in the 24 hour test incubation, 271 downregulated genes were significantly represented in both cell types, from within an array of 55,0.00 gene fragment accessions. A listing of these is shown in Table 3, in which the first column data presents the fold change against control, the second column cites the gene symbol, the third column cites the Genbank Accession, and the fourth column provides an abridged description of the gene's function.
TABLE 3: Significantly Downregulated Genes by the Action of Symadex™
MEAN
LD CHANGE GENE GENBANK
OVER SYMBOL ACCESSION DESCRIPTION
CONTROL
-17.00 ACTA2 AL713608 actin, alpha 2, smooth muscle, aorta
-9.43 " ACVRCI -" NMJ3OOO2O activin A receptor type ll-like 1
-2.07 ACYP1.. AA664719 acylphosphatase 1, erythrocyte (common) type
-21.74 ADCYAP1 NM_001117 adenylate cyclase activating polypeptide 1 (pituitary)
-12.02 ADH 1C NMJ300669 alcohol dehydrogenase 1C (class I), gamma polypeptide
-22.71 AGT angiotensinogen (serine (or cysteine) proteinase inhibitor,
NM_000029 clade A (alpha-1 antiproteinase, antitrypsin), member 8)
-2.71 ALG5 asparagine-linked glycosylatiori 5 homolog (yeast, dolichyl-
NM_013338 phosphate beta-glucosyltransferase)
-1.92 ANAPC4 NM_013367 anaphase promoting complex subunit 4
-13.15 APOA1 NM_000039 apolipoprotein A-I
-2.29 ARL6IP NM_015161 ADP-rib'osylation factor-like 6 interacting protein
-2.21 ASAH2 N-acylsphingosine amidohydrolase (non-lysosomal
AF250847 *.ceramidase) 2
-8.24 ATP1 B4 AI659245 ATPase. (Na+)/K+ transporting, beta 4 polypeptide
-9.43 ATP2B3 NM_021949 ATPase, Ca++ transporting, plasma membrane 3
-2.08 BAD NMJ304322 BCL2-aπtagoπist of cell death
-2.22 BAG2 NM_004282 BCL2-associated athanogene 2
-23.65 BBS2 T26496 Bardet-Biedl syndrome 2
-2.17 BCAP29 NMJD18844 B-cell receptor-associated protein 29
-12.57 BGN NM 001711 brglycan -1.91 BIRC5 NM_001168 bacuioviral IAP repeat-containing 5 (survivin)
-2.58 . BLM NM_000057 Bloom syndrome
-2.11 C10ORF.7 NMI006023 chromosome 10 open reading frame 7
•15.52 C3AR1 NM_004054 complement component 3a receptor 1
-1.92 CAMK1 NM_003656 calcium/calmodulin-dependent protein kinase I
•11.45 CASR BX106711 calcium-sensing receptor (hypocalciuric hypercalcemia 1 , severe neonatal hyperparathyroidism)
12.99 CCL23 NM_005064 chemokine (C-C motif) ligand 23
-2.51 CCNB2 NM_004701 cyclin B2
-2.14 CD164 NM_006016 CD164 antigen, sialomucin
-2.40 CD58 NM_001779 CD58 antigen, (lymphocyte function-associated antigen 3)
•13.25 CD5L NM_005894 CD5 antigen-like (scavenger receptor cysteine rich family)
-2.09 CDC2 NM_001786 cell division cycle 2, G1 to S and G2 to M
-2.99 CDC25C NMi.001790 cell division cycle 25C
-8.14 CDKL1 NM_004196 cyclm-dependent kinase-like 1 (CDC2-related kinase)
•19.89 CENTA1 NM_006869 centauπn, alpha 1
-2.14 CHRNA5 NM_000745 cholinergic receptor, nicotinic, alpha polypeptide 5
-2.38 CKS1B NM_001826 CDC28 protein kinase regulatory subunit 1 B
•54.48 COL1A2 NM_000089 collagen, type I, alpha 2
-2.54 COP9 constitutive photomorphogenic homolog subunit 3
COPS3 NM_003653
(Arabidopsis)
23.19 COX6A2 NM_005205 cytochrome c oxidase subunit Via polypeptide 2
-2.13 CREM - NM_001881 cAMP responsive element modulator
-2.53 CSE1L NM_001316 CSE1 chromosome segregation 1-like (yeast)
•26.26 CSRP3 NM_003476 cysteine and glycine-rich protein 3 (cardiac LIM protein)
•20.65 CYP19A1 NM_000103 cytochrome P450, family 19, subfamily A, polypeptide 1
-1.88 D8S2298E NM_005671 reproduction 8
-1.94 DCK NM_000788 deoxycytidine kinase
-3.14 DCLRE1A NM_014881 DNA cross-link repair 1A (PSO2 homolog, S. cerevisiae)
-2.09 DDX1 NM_004939 DEAD (Asp-Glu-Ala-Asp) box polypeptide 1
-2.36 DEK NM_003472 DEK oncogene (DNA binding)
-2.95 DHFR AU127142 dihydrofolate reductase
-3.39 DLEU2 NM_006021 deleted in lymphocytic leukemia, 2
-2.15 DNAJB 11 NMJ) 16306 DnaJ (Hsp40) homolog, subfamily B, member 11
-2.24 DNAJD1 NM_013238 DnaJ (Hsp40) homolog, subfamily D, member 1
■89.97 DSC3 NM_001941 desmocollin 3
-8.75 DSCR1 L1 NM_005S22 Down syndrome critical region gene 1-lιke 1
-3.07 DTYMK NM_012145 deoxythymidyiate kinase (thymidylate kinase)
-2.18 DUSP12 NMJD07240 dual specificity phosphatase 12
-2.08 DUT NM_001948 dUTP pyrophosphatase
28.71 EGFL6 NMI015507 EGF-like-domain, multiple 6
■15.24 EIF2AK4 AI630242 eukaryotic translation initiation factor 2 alpha kinase 4
-2.13 EIF2S1 NM_004094 eukaryotic translation initiation factor 2, subunit 1 alpha,
35kDa
15.36 EIF4EL3 BX111619 eukaryotic translation initiation factor 4E-like 3
-6.35 ENG BM665467 endoglin (Osler-Rendu-Weber syndrome 1)
-3.39 ERH NM_004450 enhancer of rudimentary homolog (Drosophila)
-3.32 FAIM NM_018147 Fas apoptotic inhibitory molecule
-2.11 FARS'1 NM_006567 phenylalanine-tRNA synthetase 1 (mitochondrial)
17 42 FBLN1 NM_001996 fibulin 1
■13.00 FBN 1 NM_000138 fibrillin 1 (Marfan syndrome)
11.62 FCAR NM 002000 Fc fragment of IgA, receptor for -2.87 FEN1 NM_O04111 flap structure-specific endonuclease 1
-9.02 FNTA BI715309 farnesy.ltransferase, CAAX box, alpha
-9.60 FOXN 1 NM_003593 forkhead box N1
10.14 GABRA3 NM_000808 gamma-aminobutyric acid (GABA) A receptor, alpha 3
-2.44 • GDAP1 NM_018972 ganglioside-induced differentiation-associated protein 1 gamma-glutamyl hydrolase (conjugase,
-1.96 GGH NM_003878 folylpolygammaglutamyl hydrolase)
19.73 GIPR NM_000164 gastric inhibitory polypeptide receptor
62.09 GJB5 - •NM_005268 gap junction protein, beta 5 (connexin 31.1)
-2.51 GLA NM_000169 galactosidase, alpha
-2.81 GMNN ..NM_015895 geminin, DNA replication inhibitor- guanine nucleotide binding protein (G protein), alpha
•18.16 GNAL BX 116836 activating activity polypeptide, olfactory type
•11.49 GPR1 CB992712 G protein-coupled receptor 1
•80.43 GPR15 NM_005290 G protein-coupled receptor 15
■13.80 GPR24 NM_005297 G protein-coupled receptor 24 '
-2.56 GPR54 NMJ332551 G protein-coupled receptor 54
-2.31 . H2AFX NM_002105 H2A histone family, member X
-2.35 H2AF2 NM_002106 H2A histone family, member Z
-2.63 HAT1 NM_003642' histone acetyltransferase 1
-1.97 HMGB1 NM_002128 high-mobilitygroup box 1
-2.94 HMMR NM_012484 hyaluronan-mediated motility receptor (RHAMM)
-7.42 HNF4A NM_000457 hepatocyte nuclear factor 4, alpha
-1.93 HNRPA2B1 NM 002137 heterogeneous nuclear ribonucleoprotein A2/B1
-2.08 HSGT 1 NM_007265 suppressor of S. cerevisiae gcr2
13.76 HSPB2 NM_001541 heat shock 27kDa protein 2 integrin-binding sialoprotein (bone sialoprotein, bone
•58.56 IBSP NM_004967 sialoprotein Ii)
-9.79 IL13RA2 NM__000640 interleukin 13 receptor, alpha 2
•11.23 IL1 RAP . AK095107 interleukin 1 receptor accessory protein
28.60 IL1RL1 NM 003856 interleukin 1 receptor-like 1
25.20 . IL7R NMJΪ02185 interleukin 7 receptor integrin, alpha 2b (platelet glycoprotein Hb of Ilb/Illa
•10.64 ITGA2B NM_000419 complex, antigen CD41 B)
•27,91- ITGA9 BF959890 integrin, alpha 9 integrin, alpha E (antigen CD103, human mucosal
-2.12 ITGAE NM_002208 lymphocyte antigen 1 ; alpha polypeptide)
-2.73 ITGB3BP NM_014288 integrin beta 3 binding protein (beta3-endonexin)
•15.06 ITSN1 NM_003024 intersectin 1 (SH3 domain protein) potassium inwardly-rectifying channel, subfamily J,
15.93 KCNJ12 NMJ)21012 member. 12 potassium inwardly-rectifying channel, subfamily J,
-6.83 KCNJ15 NM_002243 member 15 • potassium voltage-gated channel, KQT-like subfamily,
24.13 KCNQ2 NM_004518 member 2
-7.23 KIAA0089 NM_015141 KIAA0089 protein
-2.52 KIF2C NM_006845 kinesin family member 2C
11.04 KIF5A AL118561 kinesin family member 5A
24.24 KLRG1 NM_005810 killer cell lectin-like receptor subfamily G, member 1
-3.34 KRT 13 NM_002274 keratin 13
LC P2 lymphocyte cytosolic protein 2 (SH2 domain containing
11.85 NM_005565 leukocyte protein of 76kDa)
57.61 LIMS2 NM 017980 LIM and senescent cell antigen-like domains 2 87.48 LNX AL565198 ligand of numb-protein X
11.94 LPAAT-E NM_018361 acid acyltransferase-epsilon
-2.40 LPAAT-E NM_018361 acid acyltransferase-epsilon low density lipoprotein-related protein IB (deleted in
15.01 LRP 1 B NM_018557 tumors) • • .
12.57 LTB NM_002341 lymphotoxin beta (TNF superfamily, member 3)
-3-.14 MAD2L1 NM_002358 MAD2 mitotic arrest deficient-like 1 (yeast)
12.89 MAP6 AB058781 micrbtubule-associated protein 6
-2.14 MAPK13 NM 002754 mitogert-activated protein kinase 13
•26.53 MAPT BM714794 microtubule-associated protein tau MYC-asεociated zinc finger protein (purine-binding
-1.89 MAZ- NM_002383 transcription factor)
MCM6 minichromosome maintenance deficient 6 (MIS5
-1.97 MCM6 NM_005915 • homolog, S. pombe) (S. cerevisiae) MCM7 minichromosome maintenance deficient 7 (S.
-2.69 MCM7 NM_005916 cer'evisiae)-
-2.39 MEA NM_014623 male-enhanced antigen manηosyl (alpha-1,3-)-glycoprotein beta-1,4-N-
10.83 - MGAT4A AI364966 acetylglucosaminyltransferase, isoenzyme A
-2.20 ' MIS12 NM_024039 homolog of yeast Mis12
-2.04 MPZL1 NM_003953 myelin protein zero-like 1
-2.02 MRPL1 NM_020236 mitochondrial ribosomal protein L1
-2.97 MRPL11 NM 016050 mitochondrial ribosomal protein L11
-2.16 MRPL13 NM_014078 mitochondrial ribosomal protein L13
-2.29 MRPL23 NM_021134 mitochondrial ribosomal protein L23
-2.20 MRPL39 • NM_017446 mitochondrial ribosomal protein L39
17.46 MSLN NM_005823 mesothelin
•16.21 MT1A BM684446 metallothionein 1A (functional)
-2.09 MT2A BG505162 metallothionein 2A
•59.05 MTIF2 AI064964 I factor (complement)
-2.66 MXD3 BQ053282 MAX dimerization protein 3
21.22 MYBPC2 NM_004533 myosin binding protein C1 fast type
•50.59 MYO15A NM_016239 myosin XVA
NCF1 neutrophil cytosolic factor 1 (47kDa, chronic •
•33.67 BI 021745 granulomatous disease, autosomal 1) neutrophil cytosolic factor 2 (65kDa, chronic
■21.83 NCF2 NM_000433 granulomatous disease, autosomal 2)
-2.51 " NDUFA6 NM_002490 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 6, 14kDa
NADH dehydrogenase (ubiquinone) 1 beta subcomplex, 3,
•15.58 NDUFB3 NM_002491 12kDa
•10.52 NDUFV3 AW-139027 NADH dehydrogenase (ubiquinone) flavoprotein 3, 1OkDa
-6.89 NEB AI079911 nebulin nuclear factor of activated T-cells, cytoplasmic,
•16.69 N FATC 1 NM_006162 calcineurin-dependent 1
-2.01 NFKBIB NM_O02503 nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, beta
-2.41 NMI NM_004688 N-myc (and STAT) interactor
-2.12 NMU , NM 006681 neuromedin U
15.94 NR0B1 NM_000475 nuclear receptor subfamily 0, group B, member 1
54.82 NR2E1 NM_003269 nuclear receptor subfamily 2, group E, member 1
14.89 NR2E3 NM_016346 nuclear receptor subfamily 2, group E, member 3
•17.54 NRG1 NM 013956 neuregulin 1 -2.08 NT5C3 AA188573 5'-nuclebtidase, cytosolic III
-17.10 NT5E BM994339 δ'-nucleotidase, ecto (CD73)
-2.34 NTHL1 NM_002528 nth endonuclease Ill-like 1 (E. coli) nuclear ubiquitous casein kinase and cycliπ-dependent
-2.02 NUCKS NM_022731 kinase substrate ,
. nudix (nucleoside diphosphate linked moiety X)-type motif
-1.89 NUDT1 NM_:002452
1
-2.68 NUP107 NM_020401 nucleoporin 107kDa '
-16.33 . OLRi CD678960 lysyl. oxidase
-2.68 OXCT NMJD00436 3-oxoacid CoA transferase 1 platelet-activating factor acetylhydrolase, isoform Ib1 alpha
-12.85 PAFAH 1B1 AI674778 subunit 45kDa ■ platelet-activating factor acetylhydrolase, isofόrm Ib, beta
-2.25 PAFAH 1B2 NM_002572 subunit 3OkDa phosphoribosylaminσimidazole carboxylase,
-2.03 PAICS NMJ306452 phosphoribosylaminoimidazole succinocarboxamide synthetase
-19.65 PCDH7 NM_032457 BH-protocadherin (brain-heart)
-16.89 PCSK2 NMJD02594 • proprotein convertase subtilisin/kexin type 2
-2.21 PDCD5 NMJD04708 programmed cell death 5
-16.85 PDE11A NM_016953 phosphodiesterase 11A
-17.53 PECAM 1 BG739826 platelet/endothelial cell adhesion molecule (CD31 antigen)
.-10.55 PFKL - AK098228 phosphofructokinase, liver . .
PHAX- likely ortholog of mouse phosphorylated adaptor for RNA
-2.17 NM_032177 export
-2.95 PHF5A NM_032758 PHD finger protein 5A
-3.73 PIR51 • NM_006479 RAD51 -interacting protein
-2.39 PLK4 NM_014264 polo-like kinase 4 (Drosophila)
-8.04 PLXNA3 BF926082 plexin A3
-15.36 . PMPCB AK090763 peptidase (mitochondria! processing) beta
-1.88 POLA NM_016937 polymerase (DNA.directed), alpha
-2.62 POLE2 NM_002692 polymerase (DNA directed); epsilon 2 (p59 subunit)
-2.04 POLE4 •NM_019896 polymerase (DNA-directed), epsilon 4 (p12 subunit)
-2.24 ROLR3K polymerase (RNA) III (DNA directed) polypeptide K, 12.3
NM_016310 ' kDa
-2.49 PPIH NM_006347 peptidyl prolyl isomerase H (cyclophilin H)
-26.54 PPP1 R9A AB"033048 protein phosphatase 1 , regulatory (inhibitor) subunit 9A
-36.93 PPP2R5A AA496141 ι protein phosphatase 2, regulatory subunit B (B56), alpha isoform
-3.78 PRDM1 NM_001198 PR domain containing 1 , with ZNF domain
-3.47 PRIM1 NM_000946 primase, polypeptide 1, 49kDa
-25.92 PRLR AA708864 prolactin receptor
-22.21 PRSS21 NM_006799 protease, serine, 21 (testisin)
-18.91 PTPRG BC047734 protein tyrosine phosphatase, receptor type, G
-2.43 PTTG 1 NM_004219 pituitary tumor-transforming 1
-6.00 PXN AW969600 paxillin
-2.31 RACGAP 1 NM_013277 Rac GTPase activating protein 1
-2.28 RAD18 NMJ320165 RAD18 homolog (S. cerevisiae)
-2.21 RAD51 NM_002875 RAD51 homolog (RecA homolog, E. coli) (S. cerevisiae)
-2.04 RAD54B NM_012415 RAD54 homolog B (S. cerevisiae)
-84.42 RB1 BI769614 retinoblastoma 1 (including osteosarcoma)
-8.81 RBBP9 NM_006606 retinoblastoma binding protein 9
-7.09 RCOR1 NM 015156 REST corepressor 1 -2.02 RFC4 NM 002916 replication factor C (activator 1) 4, 37kDa
-2.31 RNASEH2A NM_006397 ribonuclease H2, large subunit
-2.29 RNF141 NM_016422 ring finger protein 141
-11.95 ROBO4 NM_019055 roundabout homolog 4, magic roundabout (Drosophila)
-2.54 RPA3 • NM_002947 replication protein A3, 14kDa
-2.22 RPC62 NMJ306468 polymerase (RNA) III (DNA directed) polypeptide C (62kD)
-39.17 RP L4 . BF308998 ribosomal protein L4
-68.37 RPS3 BM693455 ribosoma! protein S3
-2.38 RQCD1 NM_005444 RCD1 required for cell differentiationi homolog (S. pombe]
-2.43 RYR3 BU533957 ryanodine receptor 3
-2.14 SARA1 NM_020150 SARIa gene homolog 1 (S. cerevisiae)
-2.00 SCAMP3 NM_005698 secretory carrier membrane protein 3
-14.52 SCNN1G NM_001039 sodium channel, nonvoltage-gated 1, gamma sema domain, immunoglobulin domain (Ig)1 and GPI
-16.54 SEMA7A NM_003612 membrane anchor, (semaphorin) 7A
-2.84 SFRS3 NM_003017 splicing factor, arginine/serine-rich 3
-7.34 SHOX NM_000451 short stature homeobox sialyltransferase 7 ((alpha-N-acetylneuraminyl-2,3-beta-
-11.76 SIAT7A NM_018414 galactosyl-1,3)-N-acetyl galactosaminide alpha-2,6- sialyltransferase) A
-2.93 SIN3B AW051366 SIN3 homolog B, transcriptional regulator (yeast)
-2.78 SIVA NM_006427 CD27-binding (Siva) protein solute carrier family 1 (glial high affinity giutamate
-11.21 SLC1A2 NM_004171 transporter), member 2 solute carrier family 22 (organic cation transporter),
-11.51 SLC22A13 NM_004256 member 13
-18.72 SLC27A6 NMJD 14031 solute carrier family 27 (fatty acid transporter), member 6
-2.23 SLC35B1 NMJ305827 solute carrier family 35, member B1 solute carrier family 6 (neurotransmitter transporter,
-19.44 SLC6A4 NM_001045 serotonin), member 4 solute carrier family 7, (cationic amino acid transporter, y+
-27.75 SLC7A13 NM_138817 system) member 13 solute carrier family 9 (sodium/hydrogen exchanger),
-15.57 SLC9A3 NM_004174 isoform 3 solute carrier family 9 (sodium/hydrogen exchanger),
-8.20 SLC9A7 AA279477 - isoform 7
-17 45 SNAI2 NMJ303068 snail homoiog 2 (Drosophila)
-2.31 SNRPD3 NM_004175 small nuclear ribonucleoprotein D3 polypeptide 18kDa SPO11 meiotic protein covalently bound to DSB-like (S.
-14.75 SPO11 NMJ312444 cerevisiae)
-6.57 SPOCK sparc/osteonectin, cwcv and kazal-like domains
NM_004598 proteoglycan (testican)
-16.71 SPTB NM_000347 spectrin, beta, erythrocytic (includes spherocytosis, clinical type I)
-13.79 SRY NM_003140 sex determining region Y
-2.20 SSSCA1 NM_006396 Sjogren's syndrome/scleroderma autoantigen 1
-1.92 STK6 NMJ303600 serine/threonine kinase 6
-2.41 STM N 1 NMJD05563 stathmin 1/oncoprotein 18
-10.91 SULT1E1 NM_005420 sulfotransferase family 1 E1 estrogen-preferring, member 1
-293.18 SULT4A1 NM_014351 sulfotransf erase family 4A, member 1
-2.33 SUV39H2 NM_024670 suppressor of variegation 3-9 homolog 2 (Drosophila) synaptotagmin binding, cytoplasmic RNA interacting
-2.23 SYNCRIP NM_006372 protein
1481.77 SYNE1 NM 033071 spectrin repeat containing, nuclear envelope 1 12.26 TAC3> NMJD13251 tachykinin 3 (neuromedin K, neurokinin beta)
-2.04 TADA2L NMJD01488 ' transcriptional adaptor 2 (ADA2 hocn.olog,.yeast)-like '
-9.82 TCP11 NM_018679 t-complex 11 (mouse) .•• transcription factor AP-2 alpha (activating enhancer .
11.00 TFAP2A " NM_003220 binding protein 2 alpha)
20.30 TFEC NMJD12252 transcription factor EC,-
-3.41 THOC4 ' NMJ)05782 THO complex 4
•translocate of inner mitochondrial membrane 10 homolog
-2.23 TIMM 10 NM_012456
(yeast) translocase of inner mitochondrial membrane 23 homolog
-2.02 TIMM23 NM_006327 (yeast)
-2.34 TK1 NM_003258 thymidine kinase Ij soluble
-2.21 TMEM4 NM_014255 transmembrane protein 4
-2.64 TMPO H57815 thymopoietin transition protein 1 (during histone to protamine
36.20 TNP1 NM_003284 replacement)
•13.71 TPSD1 NM_012217 tryptase delta 1
-2.55 TRA2A • BF093914 transformer-2 alpha
-8.14 TRH NM_007117 thyrotropin-releasing hormone
-2.11 TSFM AW603708 Ts translation elongation factor, mitochondrial
-2.24 TTK NM_003318 TTK protein kinase
-2.13 TXNDC NM_030755 thioredoxin domain containing
•12.47 TYR P 1 NM_000550 tyrosinase-related protein 1
-1.99 U2AF1 NM_006758 U2(RNU2) small nuclear RNA auxiliary factor 1
10.12 UBL4 AA"873769 ubiquitin-like 4
-1.92 UMPK NM_012474 uridine monophosphate kinase
66.26 USP16 NM_006447 ubiquitin specific protease 16
VAMP (vesicle-associated membrane protein)-associated
12.46 VAPA AI671488 protein A, 33kDa
-2.22 VDAC 3 NM_005662 voltage-dependent anion channel 3
-2.79 VRK1 NM_003384 vaccinia related kinase 1
-2.07 " WWOX NM_016373 WW domain containing oxidoreductase zinc finger protein 145 (Kruppel-like, expressed in
30.18 ZNF145 BU607554 prornyelocytic leukemia)
-2.66 ZNF258 NM_007167 zinc finger protein 258
-2.03 ZNF265 NM_005455 zinc finger protein 265
31.24 ZNF282 NM_003575 zinc finger protein 282
-2.17 ZNRD1 NM 014596 zinc ribbon domain containing, 1
-2.31 ZW10 ZW10 homolog, centromere/kinetochore protein
NM 004724 (Drosophila)
Analysis
Review of this listing in the context of gene ontology, reveals that Symadex™ exerts a profound, if pleiotropic effect, on mechanisms of cell aggregation and proliferation and on processes associated with invasive cellular growth, -which are the hallmark of the inflammatory etiology associated with the autoimmune diseases described at the outset. More detailed analysis of the evidence in Table 3 reveals, for example, that a significant proportion of the down regulated genes are associated with mechanisms of cell surface signaling, motility, migration and adhesion, which permit inflammatory cells to cross vascular barrier and penetrate into parenchymal layers. Those practiced in the art will recognize that these ontological relationships are described more fully in literature within databases in the public domain, from which the following information has been excerpted. Those databases include DAVID (Database for Annotation, Visualization and Integrated Discovery, from the National Institute of Allergy and Infectious Disease, http://appsl.niaid.nih.gov/david/; the sister program EASE (Expression Analysis Systematic Explorer) at the same site; and the GeneCards bioinformatics project (http://genome- www.stanford.edu/genecards/index.shtml).
For example, in the differential gene expression experiment under discussion, the down regulated genes ACTA2, ACVRLl, BGN, DSC3, ENG, FBANl3 FBLNl, HMMR, IGTA2B, ITGA2B, ITGA9, ITGAE, LIMS2, LTB, MAPT, MSLN, NMI, PCDH7, PECAMl, PRDMl, SEMA7A, VAPA all participate in the regulation of these processes via direct modulation of adhesion factors, like integrins and cadherins, or by disrupting the growth factor signals that promote their expression and the assembly of accessory proteins that further facilitate the adhesion process. Of special importance in this context is the remarkable 1500 fold down-regulation of the SYNEl spectrin repeats. The accessory proteins in the nesprin family coded by this gene maintain nuclear organization and the structural integrity of the cellular cytoskeleton, Down-regulation of SYNEl would be expected to impair the ability of inflammatory cells to maintain their shape and geometry during periods of invasive motility. Thus, this effect of Symadex™ on differential gene expression of the machinery for maintaining cellular conformation would yield to the collapse of those cells during trafficking, an outcome also consistent with the histopathology of Symadex 's therapeutic mode of action.
The requisite processes for calcium ion and high energy phosphate generation are affected in tandem as evidenced by the down regulation of ATP1B4, ATP2B3, CAMKl, EGFL6, GPR24, IBSP, NUDTl, RAD54B, RYR3, and SLC9A7. Cell proliferation in turn is put in check through cell cycle blocking processes mediated by BIRC5, CCL23, CCNB2, CDC2, CDC25C, CKSlB5 CREM, EGFL6, FCAR, IL13RA2, ILlRAP, ILlRLl, MAPK13, NRGl, PTPRG5 STK6 • among other such related genes. Neuromodulation via paracrine and autocrine controls is also evident in the dqwnregulation of systems that further respond to neuroinflammatory insult, including, for example, neurotransmitter transporters associated with damaging, runaway glutamate signaling. The downregu'lated genes in this latter category are exemplified by ADCYAPl, GABRA3, GGH, KCNQ3, SLCl A2 (and its SLC family solute carrier homologs), and SULT4A1. This latter gene showed close to 300 fold down-regulation. It is a gene associated with heparan sulfation. Sulfated heparans constitute the "molecular velcro" that permits integrins to bind to laminins and thereby provide the linkage that permits invasive inflammatory cells to transmigrate through basal membranes into CNS parenchyma. Down regulation of a such a process would be expected to keep inflammatory cells within the confines of vascular cuffs, as has been observed to be the case in the histopathological evaluation on the Symadex™ treatment effect noted in Examples 2-8.
The integrated function of these genes affected by Symadex™ is consistent with the differential expression profile that has been observed with microarray experiments, as for example, in the work of Arnett HA et al., "Functional genomic ' analysis of remyelination reveals importance of inflammation in oligodendrocyte regeneration", J. Neuroscience 23(30) :9824-9832, 2003; Lindberg RLP et al, "Multiple sclerosis as a generalized CNS disease — comparative microarray analysis of normal appearing white matter and lesions in secondary progressive MS", J. Neuroimmunology 152:154-167, 2004; and Tajouri L. el al., "Quantitative and qualitative changes in gene expression patterns characterize the activity of plaques in multiple sclerosis", MoI. Brain Res. 119:170-183, 2003. These studies have cataloged, in a similar manner to the gene descriptions presented here, the characteristics of representative autoimmune inflammatory insults and subsequent recovery therefrom, especially in the context of autoimmune demyelinating models for which multiple sclerosis serves as a prime circumstance. Therefore, the assertion that the application of Symadex™ and its congeners in therapy for multiple sclerosis, and autoimmune diseases of similar etiology, is demonstrable in terms of the compound's molecular pharmacology.
. While this invention has been particularly shown and described with ' references to preferred embodiments thereof,' it will be understood by those skilled in the art that various changes in form and details may. be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims

What is claimed is:
1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000072_0001
wherein:
R is R", a hydroly sable group, or alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group, wherein Rx is -H, an optionally substituted alkyl, hydroxyl, alkoxy group, a halogen, or a group represented by the following structural formula:
Figure imgf000072_0002
or, R and R5 taken together with their intervening carbon atoms form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle; or R and R4 taken together with their intervening carbon atoms form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle;
R2 is -H, an optionally substituted Cl-ClO alkyl or an optionally substituted aryl or aralkyl or heteroaryl; R3 is -(CH2)n-NRaRb, wherein n is an integer from 1 to 5, and Ra and Rb 5 each independently are hydrogen or an optionally substituted alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry, whereiri Ry is a heteroaryl or a non-aromatic heterocycle, each optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'- substituted with C1-C4 alkyl or C1-C4 alkyl substituted with - NRcRd, wherein Rc and Rd are individually H, methyl or ethyl; and
R4, R5 and R6, are each independently -H, -OH, a halogen or an optionally substituted C1-C6 alkoxy; or
R5 and R6 taken together with their intervening carbon atoms, form a 5, 6 or 7 member, optionally substitited cycloalkyl or non-aromatic heterocycle, provided that when R is Rλ, then Ra and Rb are not H or optionally substituted alkyl and Ry is not optionally substituted N-morpholinyl, optionally substituted N-piperazinyl, or optionally substituted N-pyrazinyl.
2. The compound of Claim 1, wherein R is a hydrolysable group selected from groups (II) - (VII):
Figure imgf000073_0001
O
X V wherein
R7 and R8 are independently each H, optionally substituted C1-C6 alkyl, or optionally substituted aryl or aralkyl; R9 is carboxyl, carboxamide optionally N-substituted with C1-C4 alkyl, C1-C6 optionally, substituted alkanoyl, C1-C6 carbalkoxy, or optionally substituted aroyl;
R10 is H, C1-C6 optionally substituted alkyl or optionally substituted aryl or ar alkyl;
R11 and R12 are independently each H5 optionally substituted C1-C6 alkyl or, taken together with the atom to which they are attached, form an . optionally substituted non-aromatic heterocycle;
R13 and R14 are each independently H, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkanoyl, or optionally substituted aroyl, or, taken together, with the atom to which they are attached, form a heteroaryl or non-aromatic optionally substituted heterocycle;
R16 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, C1-C6 alkanoyl, or optionally substituted aroyl;
R21 is optionally substituted Cl-ClO alkyl, or an optionally substituted aryl or aralkyl or, R21 and R22 taken together with their intervening atoms form a 5-7 membered ring;
R22 and R23 are each independently -H, or optionally substituted Cl- C6 alkyl, provided that R22 and R23 are not simultaneously hydrogen;
R100 is optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R101 is Hi optionally substituted C1-C6 alkyl or optionally- substituted aryl or aralkyl;
R107 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, or a non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with - NRcRd; and
Q1 is O or NH.'
3. The compound of Claim 2, wherein
R10 is H, C1-C4 optionally substituted alkyl, phenyl or benzyl; R11 and R12 are independently each a H5 methyl or ethyl or, taken together with the atom to which they are attached form non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd;
R16 is a C1-C6 optionally substituted alkanoyl;
R100 is a C1-C4 optionally substituted alkyl; and group (VI) is represented by structural formulas (Via) or (VIb)
Figure imgf000075_0001
wherein
Y is a halogeri, -NO2, -NH2, -CpOH, alkyl, Cl -C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy; ring A is a 5-7 membered non-aromatic heterocycle optionally substituted at one or more substitutable ring. carbon atoms with methyl, hydroxyl, oxy, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H5 methyl or ethyl; and
R101 is H or C 1-C4 alkyl.
4. The compound of Claim 3, wherein:
R7 and R8 are each independently H or optionally substituted C1-C4 alkyl and R9 is carboxyl, carboxamide optionally N-substituted with a C1-C4 alkyl, C1-C4 alkanoyl, or C1-C4 carbalkoxy;
N, R1 ' and R12, taken together form N-pyrrolidinyl, N-piperidinyl, N- morpholinyl, N-thiomorpholinyl or N-piperazinyl, optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd 5 wherein Rc and Rd are individually H, methyl or ethyl;
R101 is H, methyl or ethyl; R16 is R161-C(O)-5 wherein R161 is a branched C3-C6 alkyl; and R107 is C1-C6 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH5 a C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy or C1-C3 alkyl sulfanyl, or -(CH2)p-(CH2)q- C(O)OH, wherein and q are independently an integer from 1 to 6. .
•The compound of Claim 4, wherein
R21 is optionally substituted Cl-ClO alkyl, phenyl or benzyl optionally substituted with a halogen, -NO2, -NH2, -COOH, alkyl, C1-C3 carbalkoxy, Cl -C 3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy; and
R22 and R23 are each independently -H, or a C 1 -C3 alkyl.
The compound of Claim 4, wherein R21 and R22, taken together with their intervening atoms, form a 5 or 6 membered non-aromatic heterocycle and R23 is -H, or a C1-C3 alkyl.
The compound of Claim 4, wherein R2 is -H, C1-C4 alkyl or C1-C4 haloalkyl, R4, R5 and R6 are each independently -H, -OH, C1-C4 alkyl or Cl- C4 haloalkyl, or R5 and R6 taken together are methylenedioxy. .
The compound of Claim -7, whe'rein
R7 and R8 are each independently H, methyl or ethyl, and R9 is a Cl ■ C4 alkanoyl;
R10 is a H, Cl -C4 alkyl; and ring A is selected from
Figure imgf000076_0001
R107 is C1-C6 alkyl or C1-C6 carboxyalkyl.
9. The compound of Claim 1 , wherein R alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group
10. The compound of Claim 9, wherein R alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is selected from:
Figure imgf000077_0001
(XVII); or R, taken together with R4 and their intervening atoms are:
(XVIII);
Figure imgf000077_0003
(χχ);
Figure imgf000077_0002
(XXI);
Figure imgf000077_0004
(XXII); and (XXIII), wherein
R17 for each occurrence, is H, optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl or heteroaryl, C1-C6 alkoxyalkyl, optionally substituted aryloxy, aralkyloxy or heteroaryloxy;
Q, for each occurrence "is O or S; and
Z is CH or N.
11. The compound of Claim 10, wherein R2 is -H, Cl -C4 alkyl or C 1 -C4 haloalkyl, R5 and R6 are each independently -H5 -OH5 C1-C4 alkyl or C1-C4 haloalkyl or Rs and R6 taken together are methylenedioxy.
12. The compounds of Claim 115 wherein R17 is H, optionally substituted C1-C6 alkyl, Cl -C6 alkoxyalkyl, or phenyl, benzyl, phenyloxy or benzyloxy, optionally substituted with one or more halogen atoms, -NO2, -NH2, -COOH, C1-C3 alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group- C1-C3 haloalkyl or C1-C3 haloalkoxy.
13. The compounds of Claim 12, wherein R17 is H3 C1-C4 alkyl, or phenyl, optionally substituted with one or more halogen atoms or C1-C3 haloalkyls.
14. The compounds of Claim 13, wherein R17 is H, C1-C4 haloalkyl, or phenyl, optionally substituted with one or more C1-C3 haloalkyls.
15. The compounds of Claim 14, wherein R17 is H, trifluoromethyl or phenyl substituted with one or more trifluoromethyls.
16. The compound of Claim 1, wherein Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry.
17. The compound of Claim 16, wherein Ry is a 5-7 member^non-aromatic -.- ■ ■• heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxy 1, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H3 methyl or ethyl.
18. The compound of Claim 16, wherein Ry is a heteroaryl.
19. The compound of Claim 18, wherein Ry is a N-pyrazinyl or N-pyridinyl.
20. ' The compound of Claim 19, wherein R2 is -H, C 1 -C4 alkyl or Cl -C4 haloalkyl and R6 is -H3 -OH5 C1-C4 alkyl or C1-C4 haloalkyl.
21. The compound of Claim 20, wherein Ry is selected form a group consisting of
Figure imgf000079_0001
wherein Q2 is CH2, NH3 or NR102, wherein R102 is methyl or ethyl.
22. The compound of Claim 1, wherein R a hydrolysable group, or R alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group.
23. The compound of Claim 22, wherein R is a hydrolysable group.
24. The compound of Claim 2*3, wherein R is selected from groups (II) - (VII):
(V);
Figure imgf000079_0002
(VI); (VII); (VIII); and
Figure imgf000079_0003
wherein
R7 and R8 are independently each H, optionally substituted C1-C6 alkyl. optionally substituted aryl or aralkyl;
R9 is carboxyl, carboxamide optionally N-substituted with C1-C4 alkyl, C1-C6 alkanoyl, C1-C6 carbalkoxy, or optionally substituted aroyl; R10 is H5 optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
Ru and R12 are independently each H, optionally substituted C1-C6 alkyl or, taken together with the atom to which they are attached, form an optionally substituted non-aromatic heterocycle;
R13 and R14 are each independently H, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkanoyl, or optionally substituted aroyl, or, taken together with the atom to which they are attached, form an optionally substituted heteroaryl or non-aromatic optionally substituted heterocycle;
R16 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, C1-C6 alkanoyl, or optionally substituted aroyl;
R21 is optionally substituted Cl-ClO alkyl, or an optionally substituted aryl or aralkyl or, R21 and R22 taken together with their intervening atoms form a 5-7 membered non-aromatic heterocycle;
R " and R are each independently -H, or a optionally substituted C1-C6 alkyl, provided that R22 and R23 are not simultaneously hydrogens;
R10 is optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R101 is H, optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R107 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, or a non-aromatic heterocycie, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with - NRcRd; and
Q1 is O or NH.
25. The compound of Claim 24, wherein Ry is an optionally substituted heteroaryl.
26. The compound of Claim 24, wherein n is an integer from 1 to 5, and Ra.and Rb, each independently are hydrogen or an alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form a 5-7 membered non- aromatic heterocycle optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or Cl-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or .ethyl.
27. The compound of Claim 26, wherein
R7 and R8 are independently each. H, optionally substituted C1-C6 alkyl or phenyl or benzyl, each optionally substituted with one or more . hydroxyl, C1-C3 alkoxy, amino, alkylamino, halogen, haloalkyl or haloalkoxy groups; and
R9 is carboxyl, carboxamide optionally N-substituted with a C1-C4 alkyl, C1-C4 alkanoyl, or C1-C4 carbalkoxy.
28. The compound of Claim 27, wherein
R10 is H or C1-C4 alkyl, phenyl or benzyl each optionally substituted with one or more hydroxyl, C1-C3 alkoxy, amino, alkylamino, halogen, haloalkyl or haloalkoxy groups;
■ R1 ' and R12 are independently each a H, methyl or ethyl or, taken together with the atom to -which they are attached form non-aromatic heterocycle, optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or Cl -C4 alkyl substituted with -NRcRd, wherein R° and Rd are individually H, methyl or ethyl;
R16 is a optionally substituted C1 -C6 alkanoyl;
RI00 is a Cl-C4 aIkyl;
R107 is C1-C6 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy or C1-C3 alkyl sulfanyl, or -(CH2)P-(CH2)q- C(O)OH, wherein and q are independently an integer from 1 to 6; and group (VI) is represented by structural formulas (Via) or (VIb)
Figure imgf000082_0001
wherein
Y is a halogen, -NO2, -NH2, -COOH5 alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy;
' ring A is a 5-7 membered non-aromatic heterocycle optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, oxy, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with Cl -C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein R0 and Rd are individually H, methyl or ethyl; and
R101 is H or C 1-C4 alkyl.
29. The compound of Claim 28, wherein either R21 is optionally substituted Cl-ClO alkyl, phenyl or benzyl optionally substituted with a halogen, -NO2, -NH2, -COOH5 alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy, and R22 and R23 are each independently -H, or a C1-C3 alkyl; or
R21 and R22, taken together with their intervening atoms, form a 5 or 6 membered non-aromatic heterocycle and R23 is -H5 or a C1-C3 alkyl.
3 O . The compound of Claim 29, wherein R2 is -H5 C 1 -C4 alkyl or C 1 -C4 haloalkyl, R4, R5 and R6 are each independently -H, -OH5 C1-C4 alkyl or Cl- C4 haloalkyl, or R5 and R6 taken together are methylenedioxy.
31. The compound of Claim 30, wherein either n is 2 or 3 and Ra and Rb, is each independently a hydrogen or a CUC3 alkyl; or
Ra and Rb 5.taken together with the nitrogen to which they are attached, form group Ry selected form a group consisting of
Figure imgf000083_0001
wherein Q2 is CH2, NH, or NR102, wherein R102 is methyl or ethyl.
32. The compound of Claim 31 , wherein:
R7 and R8 are.each independently H, methyl or ethyl, and R9 is a Cl - C4 alkanoyl;
R!O is a H, or Cl -C4 alkyl; '
NR11R12 is N-pyrrolidinyl, N-piperidinyl, N-morpholinyl, N- thiomorpholinyl or N-piperazinyl, optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl;
R101 is H5 methyl or ethyl;
R16 is a branched C3-C6 alkanoyl; and
R107 is C1-C6 alkyl or C1-C6 carboxyalkyl.
33. The compound of Claim 32, wherein ring A is selected from:
Figure imgf000083_0002
34. The compound of Claim 33, wherein R2 -H , methyl or ethyl, and R6 is -H , -OH or methyl or ethyl.
35. The compound of Claim 22; wherein R alone or taken- together with R4 5 or alternatively R5, and the intervening carbon atoms is the phenol isosteric group.
36. The compound of Claim 35, wherein the phenol isosteric group is selected from:
Figure imgf000084_0001
(XIH);
Figure imgf000084_0002
(XXII);.and
Figure imgf000084_0003
(XXIII)5 wherein r
R17 for each occurrence, is H, optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl or heteroaryl, C1-C6 alkoxyalkyl, optionally substituted aryloxy, aralkyl oxy or heteroaryl oxy;
Q is O or S; and Z is CH or N.
37. The compound of Claim 36, wherein Ry is an optionally substituted heteroaryl.
38, The compound of Claim 36, wherein n is an integer from 1 to 5, and Ra and
' Rb, each independently are hydrogen or an alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form a 5-7 member non- aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N1-
substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR°Rd, wherein Rc and Rd are individually H, methyl or ethyl.
39. ' The compound of Claim 38, wherein R2 is -H, C1-C4 alkyl or C1-C4 haloalkyl, and R5 and R6 are each independently -H, -OH, C1-C4 alkyl or C1-C4 haloalkyl, or taken together are methylenedioxy.
40. The compounds of Claim 39, wherein R17 is H, optionally substituted C1-C6 alkyl, or C1-C6 alkoxyalkyl, or phenyl, benzyl, phenyloxy or benzyloxy each optionally substituted with halogen, -NO2, -NH2, -COOH, C1-C3 alkyl, Cl- C3 carbalkoxy, C1-C3 a alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy.
41. The compound of Claim 40, wherein either n is 2 or 3 and Ra and Rb, is each independently a hydrogen or a Cl -C3 alkyl; or
Ra and Rb 5 taken together with the nitrogen to which they are ■ attached, form group Ry selected form a group consisting of
Figure imgf000085_0001
Figure imgf000086_0001
wherein Q2 is CH2, NH, or NR102, wherein R102 is methyl or ethyl.
42. The compounds of Claim 41 , wherein Rπ is H, C1-C4 alkyl, or phenyl, optionally substituted with one or more halogen atoms,, -NO2, -NH2, -COOH5 C1-C3 alkyl, C1-C3 carbalkoxy, C1-C3 a alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy.
43. The compounds of Claim 42, wherein R17 is H, C1-C4 haloalkyl or phenyl optionally substituted with one or more halogen atoms or C1-C3 haloalkyls.
44. The compound of Claim 43, wherein R2 is -H , methyl or ethyl and R6 is -H , -OH, methyl, ethyl.
45. The compounds of Claim 44, wherein R17 is H, trifluoromethyl or phenyl substituted with one or more trifluoromethyls.
46. A method of treating an inflammatory or a demyelinating disorder in a patient, comprising administering to said patient a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000086_0002
wherein: R is R", a hydrolysable group, or alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group,.
. wherein R* is -H, an optionally substituted alkyl, hydroxyl, alkoxy group, a halogen, or a group represented by the following structural formula: ' •
Figure imgf000087_0001
or, R and R5 taken together with their intervening carbon atoms form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle; or R and R4 taken together with their intervening carbon atoms form a 5, 6 or 7 member, optionally substituted, cycloalkyl or non-aromatic heterocycle; . R2 is -H, an optionally substituted optionally substituted Cl-ClO alkyl or an optionally substituted' aryl or aralkyl or heteroaryl;
. R3 is -(CH2)n-NRaRb, wherein n is an integer from 1 to 5, and Ra and
Rb, each independently are hydrogen or an optionally substituted alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry, wherein Ry is a heteroaryl or a non-aromatic heterocycie, each optionally substituted at one. or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N1- substituted with C1-C4 alkyl or C1-C4 alkyl substituted with - .NRcRd, wherein R° and Rd are. individually H, methyl or ethyl; and R4, R5 and R6, are each independently -H, -OH, a halogen or optionally substituted C1-C6 alkoxy; or R5 and R6 taken together with their intervening carbon atoms, form a 5, 6 or 7 member, optionally substitited cycloalkyl or non-aromatic heterocycle, provided that when R is Rx, then Ra and Rb are not H or optionally substituted alkyl and Ry is not optionally substituted N-morpholinyl, or optionally substituted N-pyrazinyl.
47. The method of Claim 46, wherein R is a hydrolysable group selected from groups (II) - (VII):
Figure imgf000088_0001
o
JL '^ (K), wherein
R7 and R8 are independently each H, optionally substituted C1-C6 alkyl, or optionally substituted aryl or aralkyl;
R9 is carboxyl, carboxamide optionally N-substituted with C1-C4 alkyl, optionally substituted C1-C6 alkanoyl, optionally substituted C1-C6 carbalkoxy, or optionally substituted aroyl;
R10 is H, optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
Rl ! and R12 are independently each H, optionally substituted C1-C6 alkyl or, taken together with the atom to which they are attached, form an optionally substituted non-aromatic heterocycle;
R13 and R14 are each independently H5 optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkanoyl, or optionally substituted aroyl, or, taken together with the atom to which they are attached, form a heteroaryl or non-aromatic optionally substituted heterocycle; R16 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, optionally substituted C1-C6 alkanoyl, or optionally substituted aroyl;
R21 is optionally substituted Cl-ClO alkyl, or an optionally substituted aryl or aralkyl or, R21 and R22 taken together with their intervening atoms form a 5-7 membered ring;
R22 and R23 are each independently -H, or optionally substituted Cl- C6 alkyl, provided that R22 and R23 are not simultaneously hydrogen;
R100 is optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R101 is H, optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R!07 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, or a non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with - NRcRd; and
Q1 is O or NH.
48. The method of Claim 47, wherein
R10 is H, C1-C4 alkyl, phenyl or benzyl;
Rn and R12 are independently each a H, methyl or ethyl or, taken together with the atom to which they are attached form non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd;
R16 is a optionally substituted C1-C6 alkanoyl;
Rl00 is a Cl-C4 alkyl; and group (VI) is represented by structural formulas (Via) or (VIb)
Figure imgf000089_0001
wherein
Y is a halogen, -NO2, -NH2, -COOH, alkyl, C1-C3 carbalkoxy, Cl -C3 alkoxy group, Cl -C3 haloalkyl or Cl -C3 . haloalkoxy; ring A is a 5-7 membered non-aromatic heterocycle optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, oxy, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NR0R , wherein R° and Rd are individually H, methyl or ethyl; and
R101 is H or Cl-C4 alkyl.
49. The method of Claim 48, wherein:
R7 and R8 are each independently H or Cl -C4 alkyl and R9 is carboxyl, carboxamide optionally N-substiruted with a C1-C4 alkyl, C1-C4 alkanoyl, or Cl -C4 carbalkoxy;
N, Rn and R12, taken together form N-pyrrolidinyl, N-piperidinyl, N- morpholinyl, N-thiomorpholinyl or N-piperazinyl, optionally N'-substituted -with Cl-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl; - RI0I is H, methyl or ethyU
R16 is R!61-C(O)-, wherein R161 is a branched C3-C6 alkyl; and
R107 is C1-C6 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH5 a C1-C3 alkyl, C1-C3 halo'alkyl, C1-C3 alkoxy, C1-C3 haloalkoxy or C1-C3 alkyl sulfanyl, or -(CH2)p-(CH2)q- C(O)OH, wherein and q are independently an integer from 1 to 6.
50. The method of Claim 49, wherein
R21 is optionally substituted Cl-ClO alkyl, phenyl or benzyl optionally substituted with a halogen, -NO2, -NH2, -COOH5 alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy; and
R22 and R23- are each independently -H, or a C1-C3 alkyl.
51. The method of Claim 50, wherein R21 and R22, taken together with their intervening atoms, form a 5 or 6 membered non-aromatic heterocycle and R23 is -H3 or a Cl-C3 alkyl.
52. The method of Claim 50, wherein R2 is -H5 C1-C4 alkyl or C1-C4 haloalkyl, R4, R5 and R6 are each independently -H9 -OH, C1-C4 alkyl or C1-C4 haloalkyl, or R5 and R6 taken together are methylenedioxy.
53. The method of Claim 52, wherein
R7 and R8 are each independently H5 methyl or ethyl, and R9 is a Cl- C4 alkanoyl;
R10 is a H, Cl-C4 alkyl; and ring A is selected from
Figure imgf000091_0001
R » 1'0u7/ is C1-C6 alkyl or C1-C6 carboxyalkyl.
54. The method of Claim 46, wherein R alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group
55. The method of Claim 54, wherein R alone or taken together with R4, or alternatively R5, and the intervening carbon atoms is selected from.
Figure imgf000091_0002
Figure imgf000092_0001
(XVII); or
R, taken together with R4 and their intervening atoms are:
Figure imgf000092_0002
(XVIII); (XIX); (χχ);
Figure imgf000092_0003
(XXIII)5 wherein
R17 for each occurrence, is H5 optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl or heteroaryl, C1-C6 alkoxyalkyl, optionally substituted aryloxy, aralkyloxy or heteroaryloxy;
Q3 for each occurrence, is O or S; and
Z is CH or N.
56. The method of Claim 55, wherein R2 is -H, C 1 -C4 alkyl or C 1 -CA haloalkyl, R5 and R6 are each independently -H5 -OH, C1-C4 alkyl or C1-C4 haloalkyl or R5 and R6 taken together are methylenedioxy.
57. The method of Claim 56, wherein R17 is H5 optionally substituted C 1 -C6 alkyl, C1-C6 alkoxyalkyl, or phenyl, benzyl, phenyloxy or benzyloxy, optionally substituted with one or more halogen atoms, -NO2, -NH2, -COOH5 C1-C3 alkyl, C1-C3 carbalkoxy, C1-C3 aikoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy.
58. The method of Claim 57, wherein R17 is H, C1-C4 alkyl, or phenyl, optionally substituted with one or more halogen atoms or C1-C3 haloalkyls.
59. The method of Claim 58, wherein R.!7 is H, C1-C4 haloalkyl, or phenyl, optionally substituted with one or more Cl -C3 haloalkyls.
■ 60. The method of Claim 59, wherein R17 is H, trifluoromethyl or phenyl substituted with one or more trifluoromethyls.
61. The method. of Claim 46, wherein Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry.
62. The method of Claim -61 , wherein Ry. is a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or.methoxy^and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein R0 and Rd are individually H5 methyl or ethyl.
63. The method of Claim 6I5 wherein Ry is a heteroaryl.
64. The method of Claim 63, wherein Ry is a N-pyrazinyl or N-pyridinyl.
65. The method of Claim 62, wherein R2 is -H, C1-C4 alkyl or C1-C4 haloalkyl and R6 is- -H, -OH, C 1 -C4 alkyl or Cl -C4 haloalkyl.
66. - The method of Claim 65, wherein Ry is selected form a group consisting of
; a aτnird| wherein Q2. is CH2, NH, or NR102, wherein R102 is methyl or ethyl.
67. The method of Claim 46, wherein
R a hydrolysable group, or R alone or taken together with K*, or alternatively R5, and the intervening carbon atoms is a phenol isosteric group.
68. The method of Claim 67, wherein R is a hydrolysable group.
69. The method of Claim 68, wherein R is selected from groups (II) - (VII):
Figure imgf000094_0001
(IX), wherein
R7 and Rs are independently each H, optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl;
R9 is carboxyl, carboxamide optionally N-substituted with C1-C4 alkyl, optionally substituted C1-C6 alkanoyl, optionally substituted C1-C6 carbalkoxy, or optionally substituted aroyl;
R10 is H, optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
Rπ and R12 are independently each H5 optionally substituted C1-C6 alkyl or, taken together with the atom to which they are attached, form an optionally substituted non-aromatic heterocycle;
R13 and R14 are each independently H5 optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkanoyl, or optionally substituted aroyl, or, taken together with the atom to which they are attached, form an optionally substituted heteroaryl or non-aromatic optionally substituted heterocycle; R16 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, optionally substituted C 1 -C6 alkanoyl, or optionally substituted aroyl;
R21 is optionally substituted Cl-ClO alkyl, or an optionally substituted aryl or- aralkyl or, R21 and R22 taken together with their intervening atoms form a 5-7 membered non-aromatic heterocycle;
R22 and R23 are each independently -H, or optionally substituted Cl- Cβ alkyl, provided that R22 and R23 are not simultaneously hydrogens;
R100 is optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R101 is H, optionally substituted C1-C6 alkyl or optionally substituted aryl or aralkyl;
R107 is optionally substituted C1-C6 alkyl, optionally substituted aryl or aralkyl, or a non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with - NRcRd; and
Q! is O or NH.
70. The method of Claim 69, wherein Ry is an optionally substituted heteroaryl.
71. The method of Claim 69, wherein n is an integer from 1 to 5, and Ra and Rb, each independently are hydrogen or an alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form a 5-7 membered non- aromatic heterocycle optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl.
72. The method of Claim 71, wherein ' R7 and R8 are independently each H5 optionally substituted C1-C6 alkyl or phenyl or benzyl, each optionally substituted with one or more hydroxyl, C1-C3 alkoxy, amino, alkylamino, halogen, haloalkyl or haloalkoxy groups; and
R9 is carboxyl, carboxamide optionally N-substituted with a C1-C4 alkyl, C1-C4 alkanoyl, or C1-C4 carbalkoxy.
The method of Claim 72, wherein
R10 is H, C1-C4 alkyl, or phenyl or benzyl each optionally substituted with one or more hydroxyl, C1-C3 alkoxy, amino, alkylamino, halogen, haloalkyl or haloalkoxy groups;
Ru and R12 are independently each a H, methyl or ethyl or, taken together with the atom to which they are attached form non-aromatic heterocycle, optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl;
R16 is a optionally substituted C1-C6 alkanoyl;
R)00 is a Cl-C4 alkyl;
R107 is C1-C6 alkyl optionally substituted with -OH5 -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl, C1-C3 haloalkyl, C1-C3 alkoxy, C1-C3 haloalkoxy or C1-C3 alkyl sulfanyl, or -(CH2)p-(CH2)q- C(O)OH, wherein and q are independently ah integer from 1 tθ'6; and group (VI) is represented by structural formulas (Via) or (VIb)
Figure imgf000096_0001
wherein
Y is a halogen, -NO2, -NH2, -COOH, alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy; ring A is a 5-7 membered non-aromatic heterocycle optionally substituted at one or more substitutable ring carbon atoms with methyl, hydroxyl, oxy, or methoxy, and optionally substituted at any one or more ring nitrogen atoms with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl; and
R101 is H or C 1-C4 alkyl.
74. The method of Claim 73, wherein either R21 is optionally substituted Cl-ClO alkyl, phenyl or benzyl optionally substituted with a halogen, -NO2, -NH2, -COOH, alkyl, C1-C3 carbalkoxy, C1-C3 alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy, and R22 and R23 are each independently -H, or a C1-C3 alkyl; or
R21 and R22, taken together with their intervening atoms, form a 5 or 6 membered non-aromatic heterocycle and R23 is -H, or a C1-C3 alkyl.
75. The method of Claim 74, wherein R2 is -H, Cl -C4 alkyl or Cl -C4 haloalkyl, R4, R5 and R6 are each independently -H, -OH, C1-C4 alkyl or C1-C4 haloalkyl, or R5 and R6 taken together are methylenedioxy.
76. The method of Claim 75, wherein either n is 2 or 3 and Ra and Rb, is each independently a hydrogen or a Cl-C3 alkyl; or
Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry selected form a group consisting of
Figure imgf000097_0001
wherein Q2 is CH2, NH, or NR102, wherein R102 is methyl or ethyl.
77. The method of Claim 76, wherein:
R7 and R8 are each independently H, methyl or ethyl, and R9 is a Cl - C4 alkanoyl;
.R10 is aH, or Cl -C4. alkyl;
NR11R12 is N-pyrrσlidinyl, N-piperidinyl, N-morpholinyϊ, N- •thiomorpholinyl or N-piperazinyl, optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NRcRd, wherein Rc and Rd are individually H, methyl or ethyl;
R101 is H, methyl or ethyl;
R16 is a branched C3-C6. alkanoyl; and .
R107 is C1-C6 alkyl or C1-C6 carboxyalkyl.
78. The method of .Claim 77, wherein ring A is selected from:
Figure imgf000098_0001
79. The method of Claim 78, wherein R2 -H , methyl or ethyl, and Rδ is -H , -OH or methyl or ethyl.
80. The method of Claim 67, wherein R alone or taken.together with R4, or alternatively Rs, and the intervening carbon atoms is the phenol isosteric group.
81. The method of Claim 80, wherein the phenol isosteric group is selected from: (XII);
Figure imgf000099_0001
Figure imgf000099_0002
pan);
Figure imgf000099_0003
(XVII); (XVIII); (χiχ);
Figure imgf000099_0004
(XX); (XXI); (XXII); and
Figure imgf000099_0005
(XXIII), wherein
R17 for each occurrence, is H5 optionally substituted C1-C6 alkyl, ■optionally substituted aryl or aralkyl or heteroaryl, C1-C6 alkoxyalkyl, optionally substituted aryloxy, aralkyloxy or heteroaryloxy;
Q is O or S; and
Z is CH or N.
82. The method of Claim 81 , wherein Ry is an optionally substituted heteroaryl.
83. The method of Claim 81, wherein n is an integer from 1 to 5, and Ra and Rb, each independently are hydrogen or an alkyl, or Ra and Rb, taken together with the nitrogen to which they are attached, form a 5-7 member non- aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N1- substituted with C1-C4 alkyl or C1-C4 alkyl substituted with ~NR°Rd 5 wherein Rc and Rd are individually H, methyl or ethyl.
84. The method of Claim 83, wherein R2 is -H, C 1 -C4 alkyl or C 1 -C4 haloalkyl, and R5 and R6 are each independently -H5 -OH, C1-C4 alkyl.or C1-C4 haloalkyl, or taken together are methylenedioxy.
85. The method of Claim.84, wherein R17 is H, optionally substituted C1-C6 alkyl, or C 1 -C6 alkoxyalkyl, or phenyl, benzyl, phenyloxy or benzyloxy each optionally substituted with halogen, -NO2, -NH2, -COOH5 C1-C3 alkyl, Cl- C3 carbalkoxy, C1-C3 a alkoxy group, C1-C3 haloalkyl or C1-C3 haloalkoxy.'
86. The method of Claim 85, wherein either n is 2 or 3 and Ra and Rb, is each independently a hydrogen of a Cl-C3 alkyl; or
Ra and Rb, taken together with the nitrogen to which they are attached, form group Ry selected form a group consisting of
Figure imgf000100_0001
Figure imgf000100_0002
; a anndd
Figure imgf000100_0003
. . wherein Q2 is CH2, NH, or NR102, wherein R102 is methyl or ethyl.
87. The method of Claim 86, wherein R17 is H5 C 1 -C4 alkyl5 or phenyl, optionally substituted with one or more halogen atoms, -NO2, -NH2, -COOH5 C1-C3 alkyl, C1-C3 carbalkoxy, C1-C3 a alkoxy group, Cl-C3 haloalkyl or C 1 -C3 haloalkoxy.
88. The method of Claim 87, wherein R17 is H, C 1 -C4 haloalkyl or phenyl optionally substituted with one or more halogen atoms or C1-C3 haloalkyls.
89. The method of Claim 88, wherein R2 is -H , methyl or ethyl and R6 is -H , -OH, methyl, ethyl.-
90. The method of Claim 89, wherein R17 is H, trifluoromethyl or phenyl substituted with one or more trifluoromethyls.
91. The method of Claim 46, wherein the disorder is systemic lupus,
■ inflammatory bowl disease, psoriasis, Crohn's disease, rheumatoid arthritis, sarcoid, Alzheimer's disease, , insulin dependent diabetes mellitus, . atherosclerosis, asthma, spinal cord injury, stroke, a chronic inflammatory demyelinating neuropathy, multiple sclerosis, a congenital metabolic disorder, a neuropathy with abnormal myelination, drug-induced demyelination, radiation induced demyelination, a hereditary demyelinating condition, a prion-induced demyelination,- encephalitis-induced demyelination.
92. The method of Claim 46, wherein one or more additional pharmaceutical agents is co-administered with a compound of formula (I).
93. A method of treating a patient suffering from a cancer, comprising administering to said patient a therapeutically effective amount of any of the compounds of Claims 1-45 of a pharmaceutically acceptable-salt thereof, wherein the cancer is selected from the group consisting of colorectal, ovarian, renal, sarcoma, melanoma, head & neck, hepatocellular, thyroid, non-small cell lung cancer, multidrug-resistant leukemia, lymphoma, and multiple myeloma.
94. A method of treating a patient suffering from an' acute myeloid leukemia characterized by a FLT3 mutation, comprising administering to the patient a therapeutically effective amount of a compound of Claims 1-45 or a pharmaceutically acceptable salt thereof.
PCT/US2007/003135 2006-02-08 2007-02-07 Compounds for treating inflammatory disorders, demyelinating disorders and cancers WO2007092436A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/893,375 US20080108641A1 (en) 2006-02-08 2007-08-15 Compounds for treating inflammatory disorders, demyelinating disdorders and cancers

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US77190906P 2006-02-08 2006-02-08
US60/771,909 2006-02-08
US83515306P 2006-08-02 2006-08-02
US60/835,153 2006-08-02

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/893,375 Continuation-In-Part US20080108641A1 (en) 2006-02-08 2007-08-15 Compounds for treating inflammatory disorders, demyelinating disdorders and cancers

Publications (2)

Publication Number Publication Date
WO2007092436A2 true WO2007092436A2 (en) 2007-08-16
WO2007092436A3 WO2007092436A3 (en) 2008-12-18

Family

ID=38185678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/003135 WO2007092436A2 (en) 2006-02-08 2007-02-07 Compounds for treating inflammatory disorders, demyelinating disorders and cancers

Country Status (1)

Country Link
WO (1) WO2007092436A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007143096A2 (en) * 2006-06-02 2007-12-13 Xanthus Pharmaceuticals, Inc. Compounds for treating cancers
WO2008016660A2 (en) * 2006-08-02 2008-02-07 Xanthus Pharmaceuticals, Inc. Imidazoacridine compounds for treating leukemias
WO2008016665A2 (en) * 2006-08-02 2008-02-07 Xanthus Pharmaceuticals, Inc. Imidazoacridine compounds for treating flt3 -mediated disorders
WO2009017795A1 (en) * 2007-08-02 2009-02-05 Xanthus Pharmaceuticals, Inc. Indazole compounds for treating inflammatory disorders, demyelinating disorders and cancers
WO2016174674A1 (en) * 2015-04-27 2016-11-03 The Medical Research, Infrastructure and Health Services Fund of the Tel Aviv Medical Center Egr1 targeting molecules for the treatment of inflammatory and hyperproliferative conditions

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992015583A1 (en) * 1991-03-05 1992-09-17 British Technology Group Ltd. Imidazoacridines and their antineoplastic use
WO1997038999A1 (en) * 1996-04-12 1997-10-23 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Acridone-derived compounds useful as antineoplastic and antiretroviral agents
US6229015B1 (en) * 1996-10-07 2001-05-08 Btg International Limited Acridone derivatives and method of preparation of 8-hydroxy imidazoacridinone derivatives
WO2001066545A2 (en) * 2000-03-07 2001-09-13 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 1,8-NAPHTHALIMIDE IMIDAZO[4,5,1-de]ACRIDONES WITH ANTI-TUMOR ACTIVITY
WO2006081431A2 (en) * 2005-01-28 2006-08-03 Xanthus Pharmaceuticals, Inc. Compounds for treating inflammatory and demyelinating diseases

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992015583A1 (en) * 1991-03-05 1992-09-17 British Technology Group Ltd. Imidazoacridines and their antineoplastic use
WO1997038999A1 (en) * 1996-04-12 1997-10-23 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Acridone-derived compounds useful as antineoplastic and antiretroviral agents
US6229015B1 (en) * 1996-10-07 2001-05-08 Btg International Limited Acridone derivatives and method of preparation of 8-hydroxy imidazoacridinone derivatives
WO2001066545A2 (en) * 2000-03-07 2001-09-13 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 1,8-NAPHTHALIMIDE IMIDAZO[4,5,1-de]ACRIDONES WITH ANTI-TUMOR ACTIVITY
WO2006081431A2 (en) * 2005-01-28 2006-08-03 Xanthus Pharmaceuticals, Inc. Compounds for treating inflammatory and demyelinating diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MAZERSKA ZOFIA ET AL: "C-1311: Atineoplastic" DRUGS OF THE FUTURE, BARCELONA, ES, vol. 23, no. 7, 1 July 1998 (1998-07-01), pages 702-706, XP002459524 ISSN: 0377-8282 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007143096A2 (en) * 2006-06-02 2007-12-13 Xanthus Pharmaceuticals, Inc. Compounds for treating cancers
WO2007143096A3 (en) * 2006-06-02 2008-03-27 Xanthus Pharmaceuticals Inc Compounds for treating cancers
WO2008016660A2 (en) * 2006-08-02 2008-02-07 Xanthus Pharmaceuticals, Inc. Imidazoacridine compounds for treating leukemias
WO2008016665A2 (en) * 2006-08-02 2008-02-07 Xanthus Pharmaceuticals, Inc. Imidazoacridine compounds for treating flt3 -mediated disorders
WO2008016665A3 (en) * 2006-08-02 2008-04-17 Xanthus Pharmaceuticals Inc Imidazoacridine compounds for treating flt3 -mediated disorders
WO2008016660A3 (en) * 2006-08-02 2008-04-24 Xanthus Pharmaceuticals Inc Imidazoacridine compounds for treating leukemias
WO2009017795A1 (en) * 2007-08-02 2009-02-05 Xanthus Pharmaceuticals, Inc. Indazole compounds for treating inflammatory disorders, demyelinating disorders and cancers
WO2016174674A1 (en) * 2015-04-27 2016-11-03 The Medical Research, Infrastructure and Health Services Fund of the Tel Aviv Medical Center Egr1 targeting molecules for the treatment of inflammatory and hyperproliferative conditions
AU2016255725B2 (en) * 2015-04-27 2021-09-23 The Medical Research, Infrastructure and Health Services Fund of the Tel Aviv Medical Center EGR1 targeting molecules for the treatment of inflammatory and hyperproliferative conditions

Also Published As

Publication number Publication date
WO2007092436A3 (en) 2008-12-18

Similar Documents

Publication Publication Date Title
US20080108641A1 (en) Compounds for treating inflammatory disorders, demyelinating disdorders and cancers
EP1871367A2 (en) Compounds for treating inflammatory and demyelinating diseases
CN112154142B (en) Modulators of integrated stress pathways
AU2017241837B2 (en) Substituted aminopurine compounds, compositions thereof, and methods of treatment therewith
ES2464281T3 (en) FLT3 inhibitors for immunosuppression
AU2022202181A1 (en) SMC combination therapy for the treatment of cancer
TWI653977B (en) Method for treating cancer by using 3-(4-((4-(morpholinomethyl)benzyl)oxy)-1-oxoindolin-2-yl)hexahydropyridine-2,6-dione
TWI492746B (en) Immunomodulatory compounds for treatment of lymphoma
TWI542349B (en) Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione
CA2708264C (en) Methods of using (+)-1,4-dihydro-7-[(3s,4s)-3-methoxy-4-(methylamino)-1-pyrrolidinyl]-4-oxo-1-(2-thiazolyl)-1,8-naphthyridine-3-carboxylic acid for treatment of antecedent hematologic disorders
CN116283647A (en) Prodrug modulators that integrate stress pathways
JP2010535211A (en) Indazole compounds for treating inflammatory disorders, demyelinating disorders and cancer
JP2018524347A (en) Combination therapy for the treatment of blood cancer and solid tumors
EA001379B1 (en) Method of treating a neurological disorder in a mammal, method of stimulating and promoting growth of damaged peripheral nerves, method, for promoting neuronal regeneration and growth and method for preventing neurodegeneration in a mammal
WO2012050374A2 (en) Immunotherapy for solid tumors
US9861679B2 (en) Method of treating cancer
WO2007092436A2 (en) Compounds for treating inflammatory disorders, demyelinating disorders and cancers
CN114728062A (en) Compositions and methods for improving the efficacy of immunotherapy and vaccines
CA2997671A1 (en) Combination therapies for treating cancer
JP2019147823A (en) Method of treating intracellular infection
WO2016079527A1 (en) Combination therapy
JP2017537962A (en) Solid form comprising (1E, 4E) -2-amino-N, N-dipropyl-8- (4- (pyrrolidine-1-carbonyl) phenyl) -3H-benzo [b] azepine-4-carboxamide, composition thereof And its use
Han et al. Effective prevention of lethal acute graft-versus-host disease by combined immunosuppressive therapy with prodigiosin and cyclosporine A
Hu et al. Effects of esculentoside A on autoimmune syndrome induced by Campylobacter jejuni in mice and its modulation on T-lymphocyte proliferation and apoptosis
US20230220385A1 (en) Microrna as a therapeutic agent

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 11893375

Country of ref document: US

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07763639

Country of ref document: EP

Kind code of ref document: A2