COMPOUNDS THAT INHIBIT NFkB AND BACE1 ACTIVITY

Landry; Donald W. ;   et al.

Patent Application Summary

U.S. patent application number 14/223967 was filed with the patent office on 2014-09-18 for compounds that inhibit nfkb and bace1 activity. This patent application is currently assigned to The Trustees of Columbia University in the City of New York. The applicant listed for this patent is The Trustees of Columbia University in the City of New York. Invention is credited to Shi-Xian Deng, Gangli Gong, Jeremy C. Hwang, Tae-Wan Kim, Donald W. Landry, Yidong Liu, Kirsten Alison Rinderspacher, Yuli Xie.

Application Number20140275165 14/223967
Document ID /
Family ID42129180
Filed Date2014-09-18

United States Patent Application 20140275165
Kind Code A1
Landry; Donald W. ;   et al. September 18, 2014

COMPOUNDS THAT INHIBIT NFkB AND BACE1 ACTIVITY

Abstract

The present invention relates to compounds with activity as BACE1 and NF.kappa.B modulators, and methods for treating, preventing, or ameliorating neurodegenerative diseases, such as Alzheimer's disease. The present invention is also directed to the treatment of diseases related to dysfunction of cell proliferation, the immune system and/or inflammation using such compounds or pharmaceutical compositions containing such candidate compounds.


Inventors: Landry; Donald W.; (New York, NY) ; Kim; Tae-Wan; (East Brunswick, NJ) ; Deng; Shi-Xian; (White Plains, NY) ; Gong; Gangli; (Little Neck, NY) ; Hwang; Jeremy C.; (Great Neck, NY) ; Xie; Yuli; (New York, NY) ; Liu; Yidong; (New York, NY) ; Rinderspacher; Kirsten Alison; (Bronx, NY)
Applicant:
Name City State Country Type

The Trustees of Columbia University in the City of New York

New York

NY

US
Assignee: The Trustees of Columbia University in the City of New York
New York
NY

Family ID: 42129180
Appl. No.: 14/223967
Filed: March 24, 2014

Related U.S. Patent Documents

Application Number Filing Date Patent Number
13094504 Apr 26, 2011 8685963
14223967
PCT/US2009/043011 May 6, 2009
13094504
61109891 Oct 30, 2008
61143404 Jan 8, 2009
61143532 Jan 9, 2009

Current U.S. Class: 514/314 ; 435/184; 514/311; 546/171
Current CPC Class: A61K 31/47 20130101; A61P 29/00 20180101; C07D 409/12 20130101; A61P 37/02 20180101; C07D 215/40 20130101; C07D 401/12 20130101; A61P 25/28 20180101; A61P 35/00 20180101
Class at Publication: 514/314 ; 546/171; 514/311; 435/184
International Class: C07D 215/40 20060101 C07D215/40; C07D 401/12 20060101 C07D401/12; C07D 409/12 20060101 C07D409/12

Goverment Interests



GRANT INFORMATION

[0002] This invention was made with government support under grants P50 AG08702, and 5RO1 AT001643 awarded by the National Institutes of Health, the Molecular Libraries Initiative of the National Institutes of Health Roadmap for Medical Research and the Intramural Research Program of the National Human Genome Research Institute, National Institutes of Health. The government has certain rights in the invention.
Claims



1. A composition comprising a compound of Formula I: ##STR00018## wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, and R.sup.5 are independently selected for each occurrence from the group consisting of hydrogen, halogen, alkyl, aryl, alkoxy, aryloxy, alkylthiol, arylthiol, CN, and NO.sub.2; and wherein R.sup.10 is selected from the group consisting of substituted or unsubstituted alkyl, cycloalkyl, aryl, heteroaryl and alkenyl.

2. The composition of claim 1, wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, and R.sup.5 are independently selected for each occurrence from the group consisting of: hydrogen, methyl, Cl, OCH.sub.3, CF.sub.3 and F.

3. The composition of claim 1, wherein R.sup.10 is selected from the group consisting of phenyl, 2-Nitrophenyl, 3-Nitrophenyl, 4-Nitrophenyl, 4-Methyl-2-nitrophenyl, 2-Methyl-5-nitrophenyl, 2-Nitro-4-(trifluoromethyl)phenyl, 4-Methoxy-2-nitrophenyl, 2-Methyl-5-nitrophenyl, 4-Methyl-2-nitrophenyl, 4-Methylphenyl, 2-Aminophenyl, 2-Amino-4-methyl phenyl, Thiophen-2-yl, 5-Chlorothiophen-2-yl, 5-Bromothiophen-2-yl.

4-9. (canceled)

10. A method for inhibiting the activity of a .beta.-site APP cleavage enzyme 1 (BACE1) in a cell which comprises contacting the cell with a compound of claim 1 in an amount effective to inhibit .beta.-site APP cleavage enzyme 1 activity.

11. The method of claim 10, wherein the inhibition of .beta.-site APP cleavage enzyme 1 activity reduces the metabolism of an amyloid precursor protein (APP).

12. The method of claim 10, wherein the cell is a mammalian cell.

13. The method of claim 10, wherein the cell is contacted in vitro.

14. A method for treating Alzheimer's disease in an individual, which method comprises administering to the individual an effective amount of a compound according to claim 1.

15. A method for inhibiting the activity of a Nuclear factor-.kappa. B (NF.kappa.B) in a cell which comprises contacting the cell with a compound of claim 1 in an amount effective to inhibit Nuclear factor-.kappa. B (NF.kappa.B) activity.

16. The method of claim 15, wherein the inhibition of Nuclear factor-.kappa. B (NF.kappa.B) activity reduces nuclear translocation of the Nuclear factor-.kappa. B (NF.kappa.B).

17. The method of claim 15, wherein the cell is a mammalian cell.

18. The method of claim 15, wherein the cell is contacted in vitro.

19. A method for treating a disease related to dysfunction of cell proliferation, the immune system, and/or inflammation in an individual, which method comprises administering to the individual an effective amount of a compound according to claim 1.

20. The method of claim 19, wherein the disease is cancer.

21. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 and a pharmaceutically acceptable carrier.

22. The composition of claim 1, wherein R.sup.10 is a substituted phenyl, and wherein the substituents are selected from the group consisting of hydrogen, halogen and combinations thereof.

23. The composition of claim 22, wherein the substituents are selected from the group consisting of hydrogen, fluorine, chlorine and combinations thereof.
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] The present application is a continuation of International Application No. PCT/US2009043011, filed May 6, 2009 and published in English as WO2010051064 on May 6, 2010, which claims the priority benefits of U.S. Provisional Application No. 61/109,891, filed Oct. 30, 2008; U.S. Provisional Application No. 61/143,404, filed Jan. 8, 2009; and U.S. Provisional Application No. 61/143,532, filed Jan. 9, 2009, all four of which are hereby incorporated by reference in their entireties.

I. INTRODUCTION

[0003] The present invention relates to compounds with activity as NF.kappa.B inhibitors. The present invention also relates to methods for treating, preventing, and/or ameliorating diseases related to dysfunction of cell proliferation, the immune system, inflammation and/or neurodegeneration using such compounds or pharmaceutical compositions comprising such compounds.

2. BACKGROUND OF THE INVENTION

[0004] Nuclear factor-.kappa. B (NF.kappa.B) signaling is an essential signal transduction pathway involved in inflammatory responses, oncogenesis, viral infection, the regulation of cell proliferation and apoptosis and, in particularly in the case of B and T lymphocytes, in antigenic stimulation (Ghosh, 1998, Annu. Rev. Immunol., 16, 225-260; Karin, 1999, J. Biol. Chem., 274, 27339-27342; Israel, 2000, Trends Cell. Biol., 10, 129-133; Santoro, 2003, EMBO J., 22, 2552-2560). In mammalian cells, there are five NF.kappa.B family members that dimerize: RelA, RelB, c-Rel, NF.kappa.B 2/p100/p52 and NF.kappa.B 1/p105/p50. NF.kappa.B, whose predominant form is a heterodimeric transcription factor composed of p50 and RelA subunits, remains sequestered in the cytoplasm through association with members of an inhibitory family of proteins known as I.kappa.B. Upon stimulation by the cytokines TNF-.alpha. and interleukin-1, endotoxin (LPS), microbial and viral infections, pro-inflammatory signals converge on the canonical IkB kinase complex (IKK), a protein complex that is composed of two kinase subunits, IKK.alpha./IKK-1 and IKK.beta./IKK-2 and a structural/regulatory subunit NEMO/IKK-.gamma.. Once activated IKK complex phosphorylates IkB proteins, triggering their ubiquitination and subsequent degradation by the proteasome. Free NF.kappa.B can then move into nucleus to initiate or up-regulate gene expression.

[0005] Although IKK.alpha. and IKK.beta. exhibit striking structural similarity (52%), genetic studies have shown that they are involved in two pathways for the activation of NF.kappa.B (Pomerantz, 2002, Molecular Cell 2002 10: 693-695). IKK.beta. has been identified as the pro-inflammatory kinase responsible of activation of classical NF.kappa.B complexes, whereas IKK.alpha. in association with NF.kappa.B inducing kinase (NIK) plays an essential role in the non-canonical NF.kappa.B signaling pathway (Senftleben, 2001, 293: 1495-1499).

[0006] NF.kappa.B plays an essential role in the development and progression of cancer, including breast cancer. Animal studies suggest the presence of constitutively active NF.kappa.B at an early stage during neoplastic transformation of mammary cells (Clarkson et al., 2000, J Bio Chem. 275(17):12737-42). NF.kappa.B inhibits apoptosis in mouse mammary epithelia (Soy ak et al., 1999, Cell Growth Differ. 10(8):537-44) and selective activation of NF.kappa.B subunits have been found in human breast cancer cell lines and patient samples (Sovak et at, 1997, J Clin Invest. 100(12):2952-60; Cogswell et al., 2000, Oncogene 19(9):1123-31). An inverse correlation between the levels of NF.kappa.B activation and estrogen receptor expression has been reported (Nakshatri et al., 1997, Mol Cell Biol. 17(7):3629-39) and inhibition of NF.kappa.B in breast cancer cells induces spontaneous apoptosis (Sovak et al., 1999, Cell Growth Differ. 10(8):537-44; Cogswell et al., 2000, Oncogene 19(9):1123-31). Paclitaxel-induced sensitivity of breast cancer cell lines was enhanced by an NF.kappa.B inhibitor, parthenolide (Patel et al., 2000, Oncogene 19(36):4159-69; Newton et al., 1999, J Bio Chem. 274(26):18827-35). The Mullerian inhibiting substance was also found to inhibit breast cancer growth through NF.kappa.B mediated pathway (Segev et al., 2000, J Bio Chem. 275(37):28371-9). Furthermore, the transactivation function of NF.kappa.B is negatively regulated by IKI411 in breast cancer cell lines (Newton et al., 1999, J Bio Chem. 274(26):18827-35). Lastly, overexpression of HER2neu can activate NF.kappa.B through the activation of Akt pathway and block apoptosis (Zhou et al., 2000, J Bio Chem. 275(10:8027-31). All these reports together suggest that NF.kappa.B plays an important role in cancer generally and in breast cancer specifically.

[0007] In light of the foregoing, inhibition of NF.kappa.B activation represents a target for development of new anti-inflammatory and anti-cancer drugs (Poulaki, 2002, Am J Pathol. 161: 2229-2240). Among many protein actors in NF.kappa.B signaling pathway, IKK complex represents one of the most promising molecular targets for discoveries of new specific NF.kappa.B inhibitors. To minimize the potential toxicity effects in vivo, therapeutic success will greatly depend on the abilities of the NF.kappa.B inhibitors to block activating signals without modifying the basal level of NF.kappa.B activity. For example, May et al. described a cell-permeable peptidic inhibitor that specifically blocks the pro-inflammatory NF.kappa.B activation by disrupting the constitutive NEMO interaction with IKK kinases (May, 2000, Science 289, 1550-1554; May, 2002, J. Biol. Chem. 277, 45992-46000). Accordingly, it would be desirable to identify additional small molecules that can reduce NF.kappa.B activity by selectively inhibiting NF.kappa.B and/or components of the NF.kappa.B signaling pathway.

3. SUMMARY OF THE INVENTION

[0008] The present invention relates to compounds which inhibit NF.kappa.B activity. The compounds of the invention may be used to inhibit NF.kappa.B activity in a subject, or in a cell in culture.

[0009] The present invention also provides a method for the treatment of a dysfunction of cell proliferation, the immune system and/or inflammation in an individual, wherein the dysfunction is associated with NF.kappa.B activity, by administering to an individual in need of such treatment a pharmaceutical composition comprising at least one compound of Formulas I-III in an amount effective to treat the dysfunction.

[0010] In particular non-limiting embodiments, the present invention relates to a compound of Formula I:

##STR00001##

and to salts, esters and prodrugs of the compounds of Formula I. Additionally, the present invention describes methods of synthesizing and using compounds of Formula I.

[0011] In other non-limiting embodiments, the present invention relates to a compound of Formula II:

##STR00002##

and to salts, esters and prodrugs of the compounds of Formula II. Additionally, the present invention describes methods of synthesizing and using compounds of Formula II.

[0012] In further non-limiting embodiments, the present invention relates to a compound of Formula III:

##STR00003##

and to salts, esters and prodrugs of the compounds of Formula III. Additionally, the present invention describes methods of synthesizing and using compounds of Formula III.

[0013] The present invention also provides a method of inhibiting the activity of NF.kappa.B, by contacting the NF.kappa.B, an NF.kappa.B associated protein, or a cell expressing NF.kappa.B, with at least one compound of Formulas I-III (meaning Formula I, Formula II, or Formula III) in an amount effective to inhibit the activity of NF.kappa.B.

[0014] In one non-limiting embodiment, the NF.kappa.B is expressed by a cell, for example, a mammalian cell, and the cell is contacted with an effective amount of at least one compound of Formulas I-III.

[0015] In another embodiment, the NF.kappa.B, or cell expressing NF.kappa.B, is contacted with an effective amount of at least one compound of Formulas I-III in vitro.

[0016] In other non-limiting embodiments, the pharmaceutical composition may optionally be used in conjunction with one or more additional compound for the treatment of a dysfunction of cell proliferation, the immune system and/or inflammation.

[0017] It has further been discovered that some of these compounds inhibit BACE1, an enzyme that participates in the production of A.beta.. As A.beta. has been linked to neurodegeneration and to Alzheimer's Disease in particular, such compounds may be used in the treatment of neurodegenerative conditions and in particular in Alzheimer's Disease and related conditions.

4. BRIEF DESCRIPTION OF THE DRAWINGS

[0018] FIG. 1A-B depicts general structure of N-(quinolin-8-yl)benzenesulfonamides. A) Open chain and B) cyclized structures.

[0019] FIG. 2 depicts a method for synthesizing a particular compound of Formula II (the particular compound identified as compound 9) and a particular compound of formula III (the particular compound identified as compound I).

[0020] FIG. 3A-B depicts (A) a one-pot synthesis reaction mechanism synthesizing compound 9a of Formula II and compound I a of Formula III. (B) depicts the synthesis of particular compounds of Formula II (the particular compounds identified as 9i, 9j, and 9k).

[0021] FIG. 4 depicts a one-pot synthesis reaction mechanism for converting particular compounds of Formula II into particular compounds of Formula III. The particular R groups of the starting compounds (9a-p) and the corresponding synthesized compounds (1a-p) are identical in each synthesis, and are illustrated in the table along with the individual yields of compounds 1a-p.

[0022] FIG. 5A-B depicts (A) The two-color dual luciferase based assay. I.kappa.B.alpha. is fused to a luciferase emitting greenlight (CBG68), while a red light-emitting luciferase (CBR) is present in its native state. Both luciferases are regulated by binding of the tet repressor to the tet operator (TO) binding site (TO), allowing simultaneous induction of the luciferases by the CMV promoter upon addition of doxycycline and test compounds. The CBR protein serves to normalize for non-specific effects, while the levels of the I.kappa.B.alpha.-CBG68 protein increase uniquely with inhibition of the NF.kappa.B pathway leading to I.kappa.B.alpha. degradation. (B) The nuclear translocation assay. TNF.alpha. stimulation results in nuclear translocation of NF.kappa.B, which is detected by immunofluorescence using an anti-p65 antibody and a secondary antibody labeled with an Alexa 647 fluorophore (A647).

[0023] FIG. 6 depicts several compounds identified by the NFkB inhibition screens illustrated in FIG. 4, including Structures of MG-132 (1), BAY11-7082 (2), a quinazoline based inhibitor of AP1 and NF.kappa.B mediated transcription (3), N-(quinolin-8-yl)benzenesulfonamide; PubChem CID: 161167 (4), and C7-locked N-(quinolin-8-yebenzenesulfonamide (5); PubChem CID: 659101.

[0024] FIG. 7A-D depicts the NF.kappa.B inhibitory effect achieved by exemplary compounds of Formulas I and III in three cell-based NF.kappa.B activity assays. FIGS. 7A and C show the inhibitory effect of the compounds as measured by an I.kappa.B.alpha. stabilization assay, NF.kappa.B nuclear-translocation assay, and NF.kappa.B-bla expression assay. In the I.kappa.B.alpha. stabilization assay, a higher efficacy of I.kappa.B.alpha. stabilization compared to stabilization achieved by MG-132 (an I.kappa.B.alpha. stabilizing agent) indicates greater NF.kappa.B inhibition. I.kappa.B.alpha. stabilization was measured as an increase in luminescence of a green luciferase I.kappa.B.alpha. construct. Stabilization at a lower EC.sub.50 concentration further indicates more potent NF.kappa.B inhibition. A red luciferase under the same regulatory control as the I.kappa.B.alpha. construct was used as a control to monitor cell uniformity and non-specific effects. Compounds that stabilized I.kappa.B.alpha. were those that increased green luminescence with minimal effects on red luminescence. (.sup.aEC.sub.50 values from the I.kappa.B.alpha. stabilization assay shown for the green luminescence reporter along with the % efficacy. .sup.bEC.sub.50 values from the ratio of the green and red luminescent values for the I.kappa.B.alpha. stabilization. Data are averages from two to three experiments where each experiment consisted of concentration-titration for each compound performed in duplicate and fitting concentration-response curves to the response after bioassay. NA, not applicable; the compound only showed a strong inhibitory response in the original qHTS (IC.sub.50=2.5 .mu.M, 95% inhibition) in the non-specific (red luminescence) dataset.)

[0025] In the nuclear-translocation and NF.kappa.B-bla expression assays, inhibition of NF.kappa.B nuclear-translocation or NF.kappa.B-bla expression at a lower IC.sub.50 concentration indicates a stronger NF.kappa.B inhibitory effect. Cytotoxicity for each compound is also shown. (.sup.cIC.sub.50 values are derived from curve-fitting to data from a single experiment performed in triplicate. ND, not determined. All compounds showed >90% efficacy in the translocation assay except compounds 15 (78%) and 27 (75%). The cytotoxicity assay was performed in OCI-Ly3 cells using a 4 h endpoint.) FIG. 7B shows the synthetic scheme used to generate compounds 6-18 and 23-28 of Formula I. FIG. 7D shows the synthetic scheme used to generate compounds 19-22 of Formula I and compounds 28-40 of Formula III.

[0026] FIG. 8 depicts a sAPP.beta. assay utilized to identify inhibitors of BACE1. The assay utilizes SY5Y cells stably overexpressing BACE-GFP and SEAP-APPwt. Cells are incubated with a candidate compound for 6 hours prior to harvesting media. BACE1-mediated cleavage of APP results in the release of sAPP.beta. into the media. .alpha.-secretase, a competing enzyme that is non-amyloidogenic, also cleaves APP to release sAPP.beta.. Using an sAPP.beta.-specific antibody (s.beta.wt), SEAP-sAPP.beta. is specifically captured in a modified ELISA assay. Detection is achieved by addition of the fluorescent alkaline phosphatase substrate, 4-methylumbelliferyl phosphate (4-MUP).

[0027] FIG. 9A-B depicts a targeted screen of N-(quinolin-8-yl)benzenesulfonamides, a small-scale screen using 176 structurally-related compounds based on the scaffold of N-(quinolin-8-yl)benzenesulfonamides. A) Primary screen was conducted in 96-well format at 2 .mu.M concentration in duplicate. Data points represent mean of two determinations. The Z' factors for the four 96-well plates used in the screen were all above 0.7, B) Statistics from the primary screen. The threshold value for hit selection was set at 3 standard deviations, or 59.76%, yielding 6 hit compounds and a hit rate of 3.41%.

[0028] FIG. 10A-B depicts SAR studies of N-(quinolin-8-yl)benzenesulfonamides. 28 full and partial hits, shown in (A) for compounds 1-26 and (B) for compounds 27 and 28, from the targeted screen of N-(quinolin-8-yl)benzeriesulfonamides were characterized in SY5Y-BACEGFP-SEAPAPPwt cells using the cell-based BACE1 assay. All compounds were tested at 8 concentrations (30, 10, 3, 1, 0.3, 0.1, 0.03, and 0.01 .mu.M), and data points were analyzed with Origin software and fitted using a logistic model for IC.sub.50 determination.

[0029] FIG. 11A-B depicts Lentiviral-mediated transduction of APPsw in primary neurons. Primary cortical neurons were harvested from wild-type P0 mice and cultured according to established protocols. Lentivirus harboring human Swedish mutant APP (Lenti-APPsw) was packaged using ViraPower Lentiviral Packaging mix (Invitrogen) according to the manufacturer's protocol. DIV-14 neurons were incubated for 24 hours with primary culture media containing the indicated volume (in .mu.l) of virus (LV-1 and LV-2 denote 2 separate batches of virus, 0 denotes no virus was used). After infection, neurons were incubated for 72 hours with primary culture media. Media was collected for A.beta.40 ELISA (BioSource), and cell lysates were probed with APPCT antibody to visualize transduced full-length APP. (A) Bar graph depicting results; (B) Assay results.

[0030] FIG. 12 depicts immunocytochemistry of primary cortical neurons. DIV-14 primary cortical neurons from wild-type mice were fixed, permeabilized, and stained with TUT I to visualize neuronal .beta.-tubulin (green). Cell nuclei (blue) were visualized with a DAPI co-stain. Cells were imaged with the Nikon C1 digital confocal system at 60.times. magnification.

[0031] FIG. 13A-B depicts the effect of CU-264 on A.beta. expressing primary cortical neuron cultures from wild-type mice transformed with lentivirus harboring APPsw. CU-264, which exhibited sub-micromolar potency for sAPP.beta. reduction in SY5Y-BACEGFP-SEAPAPPwt cells, does not reduce A.beta..sub.40 levels in primary neurons when the cells are incubated with 2 .mu.M CU-264. At 10 .mu.M, CU-264 increased A.beta. levels. (A) Bar graph depicting results; (B) Assay results.

5. DETAILED DESCRIPTION

[0032] For clarity and not by way of limitation, this detailed description is divided into the following sub-portions:

[0033] (i) definitions;

[0034] (ii) synthesis schemes;

[0035] (iii) methods of treatment; and

[0036] (iv) pharmaceutical compositions.

5.1 Definitions

[0037] The terms used in this specification generally have their ordinary meanings in the art, within the context of this invention and in the specific context where each term is used. Certain terms are discussed below, or elsewhere in the specification, to provide additional guidance to the practitioner in describing the compositions and methods of the invention and how to make and use them.

[0038] In one non-limiting embodiment, the NF.kappa.B is a human NF.kappa.B. The NF.kappa.B is preferably, but not by way of limitation, encoded by the Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NF.kappa.B1) (GenBank accession number NM.sub.--003998), Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (p49/p100) (NF.kappa.B2) (GenBank accession numbers NM.sub.--001077493, NM.sub.--001077494, or NM.sub.--002502), Homo sapiens v-rel reticuloendotheliosis viral oncogene homolog A (avian) (RELA) (GenBank accession number NM.sub.--021975), Homo sapiens v-rel reticuloendotheliosis viral oncogene homolog B (RELB) (GenBank accession number NM.sub.--006509), or Homo sapiens v-rel reticuloendotheliosis viral oncogene homolog (avian) (REL) (GenBank accession number NM.sub.--002908), or any nucleic acid that encodes a human NF.kappa.B polypeptide. Alternatively, NF.kappa.B can be encoded by any nucleic acid molecule exhibiting at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or up to 100% homology to any one of the NF.kappa.B genes (as determined by standard software, e.g. BLAST or FASTA), and any sequences which hybridize under stringent conditions to these sequences, which retain NF.kappa.B activity.

[0039] In other non-limiting embodiments, an NF.kappa.B of the invention may be characterized as having an amino acid sequence described by GenBank accession numbers: NP.sub.--003989, NP.sub.--001070961, NP.sub.--001070962, NP.sub.--002493, NP.sub.--068810, NP.sub.--006500, and NP.sub.--002899, or any other amino acid sequence at least 90% homologous thereto which retains NF.kappa.B activity.

[0040] In another embodiment, NF.kappa.B comprises a family of proteins which includes, but is not limited to, the proteins NF.kappa.B1 (p50), NF.kappa.B2 (p52), RelA (p65), RelB and c-Rel.

[0041] The BACE1, NF.kappa.B or APP may be a recombinant BACE1, NF.kappa.B or APP polypeptide encoded by a recombinant nucleic acid, for example, a recombinant DNA molecule, or may be of natural origin.

[0042] The term "dysfunction of the immune system" refers to any irregular activation of the innate and adaptive immune response associated with an increase in NF.kappa.B activity, for example, aberrant T-cell or B-cell development, maturation, and/or proliferation.

[0043] The term "inflammation" encompasses both acute responses (i.e., responses in which the inflammatory processes are active) as well as chronic responses (i.e., responses marked by slow progression and formation of new connective tissue). In certain non-limiting embodiments, a disease associated with inflammation that is to be treated by a compound of the instant invention is, by way of example, but not by way of limitation, type I hypersensitivity, atopy, anaphylaxis, asthma, osteoarthritis, rheumatoid arthritis, septic arthritis, gout, juvenile idiopathic arthritis, still's disease, ankylosing spondylitis, inflammatory bowel disease, Crohn's disease or inflammation associated with vertebral disc herniation.

[0044] According to the invention, a "subject" or "patient" is a human or non-human animal. Although the animal subject is preferably a human, the compounds and compositions of the invention have application in veterinary medicine as well, e.g., for the treatment of domesticated species such as canine, feline, and various other pets; farm animal species such as bovine, equine, ovine, caprine, porcine, etc.; wild animals, e.g., in the wild or in a zoological garden; and avian species, such as chickens, turkeys, quail, songbirds, etc.

[0045] The term `alkyl` refers to a straight or branched C.sub.1-C.sub.20, preferably C.sub.1-C.sub.5, hydrocarbon group consisting solely of carbon and hydrogen atoms, containing no unsaturation, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl).

[0046] The term "alkenyl" refers to a C.sub.2-C.sub.20, preferably C.sub.1-C.sub.5, aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be a straight or branched chain, e.g., ethenyl, 1-propenyl, 2-propenyl (allyl), iso-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl

[0047] The term "cycloalkyl" denotes an unsaturated, non-aromatic mono- or multicyclic hydrocarbon ring system such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl. Examples of multicyclic cycloalkyl groups include perhydronapththyl, adamantyl and norbornyl groups bridged cyclic group or sprirobicyclic groups, e.g., Spiro (4,4) non-2-yl.

[0048] The term "aryl" refers to aromatic radicals having in the range of about 6 to about 14 carbon atoms such as phenyl, naphthyl, tetrahydronapthyl, indanyl, biphenyl.

[0049] The term "heterocyclic" refers to a stable 3- to 15-membered ring radical which consists of carbon atoms and one or more, for example, from one to five, heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For purposes of this invention, the heterocyclic ring radical may be a monocyclic or bicyclic ring system, which may include fused or bridged ring systems, and the nitrogen, carbon, oxygen or sulfur atoms in the heterocyclic ring radical may be optionally oxidized to various oxidation states. In addition, a nitrogen atom, where present, may be optionally quaternized; and the ring radical may be partially or fully saturated (i.e., heteroaromatic or heteroaryl aromatic).

[0050] The heterocyclic ring radical may be attached to the main structure at any heteroatom or carbon atom that results in the creation of a stable structure.

[0051] The term "heteroaryl" refers to a heterocyclic ring wherein the ring is aromatic.

[0052] The substituents in the `substituted alkyl`, `substituted alkenyl`, `substituted cycloalkyl`, `substituted aryl,` `substituted heteroaryl` `substituted alkoxy,` `substituted aryloxy,` `substituted alkylthiol,` and `substituted arylthiol` may be the same or different, with one or more selected from the groups hydrogen, halogen, acetyl, nitro, oxo (.dbd.O), CF.sub.3, NH.sub.2, OCH.sub.3, or optionally substituted groups selected from alkyl, alkoxy and aryl.

[0053] The term "halogen" refers to radicals of fluorine, chlorine, bromine and iodine.

[0054] The term "BACE 1" refers to a polypeptide which mediates the cleavage of APP in the .beta.-amyloidgenic pathway, producing an sAPP.beta. ectodomain APP metabolite, which is released into the extracellular space, and an intracellular C-terminal fragment (CTF). In one non-limiting embodiment, the BACE1 is a human BACE1. The BACE1 is preferably encoded by the Homo sapiens beta-site APP-cleaving enzyme 1 (BACE1) gene (GenBank accession numbers NM.sub.--012104, NM.sub.--138972, NM.sub.--138971, or NM.sub.--138973), or any nucleic acid which encodes a human BACE1 polypeptide. Alternatively, BACE1 can be encoded by any nucleic acid molecule exhibiting at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or up to 100% homology to a BACE1 gene (as determined by standard software, e.g. BLAST or FASTA), and any sequences which hybridize under stringent conditions to these sequences which retain BACE1 activity, where stringent conditions are as described in U.S. Published Patent Application US20030082140, which is hereby incorporated by reference in its entirety and for all purposes.

[0055] In other non-limiting embodiments, a BACE1 of the invention may be characterized as having an amino acid sequence described by GenBank accession numbers: NP.sub.--036236, NP.sub.--620428, NP.sub.--620427 and NP.sub.--620429, or any other amino acid sequence at least 90%, or at least 95% homologous thereto, which retains BACE1 activity.

[0056] The terms "APP" or "amyloid precursor protein" refers to a substrate of BACE1 which may be metabolized into an ectodomain sAPP.beta. fragment and a C-terminal fragment (CTF). In one embodiment, APP is an integral membrane protein expressed in many tissues and concentrated in, for example, the synapses of neurons. In one non-limiting embodiment, APP is a human APP, for example, Homo sapiens amyloid beta (A4) precursor protein (APP) encoded by an APP gene (e.g., GenBank Accession numbers: NM.sub.--201414, NM.sub.--201413, or NM.sub.--000484), or any nucleic acid that encodes a human APP polypeptide. Alternatively, APP can be encoded by any nucleic acid molecule exhibiting at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or up to 100% homology to any one of the APP genes (as determined by standard software, e.g. BLAST or FASTA), and any sequences which hybridize under stringent conditions to these sequences.

[0057] In other non-limiting embodiments, APP may be characterized as comprising an amino acid sequence described by GenBank accession numbers: NP.sub.--958817, NP.sub.--958816, or NP.sub.--000475, or any other amino acid sequence at least 90% or at least 95% homologous thereto and is cleavable by a human BACE1 protein. In non-limiting embodiments APP may be comprised in a fusion protein.

5.2 Synthesis Schemes

[0058] The present invention provides for compounds that inhibit the activity of NFkB and/or BACE1.

[0059] In certain non-limiting embodiments, the invention provides for compounds of the following Formula

##STR00004##

[0060] wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5 are independently selected for each occurrence from the group consisting of hydrogen, halogen, alkyl, aryl, alkoxy, aryloxy, alkylthiol, arylthiol, CN, and NO.sub.2; and

[0061] wherein R.sup.10 is selected from the group consisting of substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl and substituted or unsubstituted alkenyl.

[0062] In non-limiting embodiments, R.sup.1, R.sup.2, R.sup.3, R.sup.4 and R.sup.5 are independently selected for each occurrence from the group consisting of hydrogen, methyl, Cl, OCH.sub.3, CF.sub.3, Br, and F. In a specific non-limiting embodiment, R.sup.4 and R.sup.5 are hydrogen, and R.sup.1, R.sup.2, and R.sup.3 are independently hydrogen, halogen, alkyl, aryl, CN, alkoxy, aryloxy, NO.sub.2, alkylthio, and arylthio.

[0063] In other preferred embodiments, R.sup.10 is selected from the group consisting of phenyl, naphthyl, 2-Nitrophenyl, 3-Nitrophenyl, 4-Nitrophenyl, 4-Methyl-2-nitrophenyl, 2-Methyl-5-nitrophenyl, 2-Nitro-4-(trifluoromethyl)phenyl, 4-Methoxy-2-nitrophenyl, 2-Methyl-5-nitrophenyl, 4-Methyl-2-nitrophenyl, 4-Methylphenyl, 2-Aminophenyl, 2-Amino-4-methyl phenyl, Thiophen-2-yl, 5-Chlorothiophen-2-yl, 5-Bromothiophen-2-yl.

[0064] In one non-limiting embodiment, compounds of Formula I may be synthesized according to the following scheme:

##STR00005##

wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5 and R.sup.10 are defined as above for Formula I.

[0065] In other non-limiting embodiments, compounds of Formula I may be synthesized by any means known in the art.

[0066] In another non-limiting embodiment, the compound defined by Foiinula I is:

##STR00006##

[0067] In other non-limiting embodiments, the compound defined by Formula I is:

TABLE-US-00001 A ##STR00007## Compound R1 R2 R3 IC.sub.50 (sAPP.beta.) (.mu.M) CU-131 1-naphthalene H H 6.46 CU-138 1-naphthalene Br H 9.51 CU-150 2-thiophene H H 8.03 CU-151 4-methoxyphenyl H H 5.92 CU-152 4-nitrophenyl H H 4.63 CU-153 2-nitrophenyl H H 1.58 CU-155 2-naphthalene H H 2.20 CU-156 8-quinoline H H 4.23 CU-163 2-naphthalene Br H 0.79 CU-166 phenyl Br H 13.45 CU-167 4-methylphenyl Br H 1.09 CU-168 8-quinoline Br H 17.17 CU-225 4-methylphenyl H Me 3.79 CU-242 2-aminophenyl H H 3.82 CU-261 4-methoxy-2-nitrophenyl H H 1.92 CU-264 4-trifluoromethyl-2-aminophenyl H H 0.53 CU-265 4-trifluoromethyl-2-nitrophenyl H H 2.95 CU-268 2-nitrophenyl OMe H 8.11 CU-271 4-methoxy-2-aminophenyl H H 2.74 CU-273 2-nitrophenyl H Me 12.43 CU-274 2-aminophenyl H Me 11.11 CU-278 4-methoxy-2-aminophenyl H Me 11.48 CU-280 4-trifluoromethyl-2-nitrophenyl H Me 5.51 CU-281 4-trifluoromethyl-2-aminophenyl H Me 3.28 CU-294 2-naphthalene OMe H 3.75 CU-295 1-naphthalene OMe H 3.63 B Compound Structure IC.sub.50 (sAPP.beta.) (.mu.M) CU-262 ##STR00008## 0.82 CU-282 ##STR00009## 1.11

[0068] In other non-limiting embodiments, the invention provides for compounds of the following Formula II:

##STR00010##

[0069] wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are independently selected for each occurrence from the group consisting of hydrogen, halogen, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted alkoxy, substituted or unsubstituted aryloxy, substituted or unsubstituted alkylthiol, substituted or unsubstituted arylthiol, CN, and NO.sub.2. In a specific non-limiting embodiment, R.sup.4 and R.sup.5 are hydrogen, and R.sup.1, R.sup.2, and R.sup.3 are independently hydrogen, halogen, alkyl, aryl, CN, alkoxy, aryloxy, NO.sub.2, alkylthio, and arylthio.

[0070] In a non-limiting embodiment, R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are independently selected for each occurrence from the group consisting of hydrogen, methyl, Cl, OCH.sub.3, CF.sub.3 and F.

[0071] In another non-limiting embodiment, the compound defined by Formula II is:

##STR00011##

[0072] In another non-limiting embodiment, compounds of Formula II may be synthesized according to the following scheme:

##STR00012##

wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are defined as above for Formula II.

[0073] In other non-limiting embodiments, compounds of Formula II may be synthesized by any means known in the art.

[0074] In other non-limiting embodiments, the compounds of Formula II may be synthesized according to the following scheme:

##STR00013##

wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are defined as above for Formula II.

[0075] In other non-limiting embodiments, the invention provides for compounds of the following Formula III:

##STR00014##

[0076] wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are independently selected for each occurrence from the group consisting of hydrogen, halogen, alkyl, aryl, alkoxy, aryloxy, alkylthiol, arylthiol, CN, and NO.sub.2.

[0077] In non-limiting embodiments, R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8, and R.sup.9 are independently selected for each occurrence from the group consisting of hydrogen, methyl, Cl, OCH.sub.3, CF.sub.3, OH, and F. In a specific non-limiting embodiment, R.sup.4 and R.sup.5 are hydrogen, and R.sup.1, R.sup.2, and R.sup.3 are independently hydrogen, halogen, alkyl, aryl, CN, alkoxy, aryloxy, NO.sub.2, alkylthio, and arylthio.

[0078] In non-limiting embodiments, compounds of Formula III may be synthesized according to the following scheme:

##STR00015##

wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are defined as above for Formula III.

[0079] In other non-limiting embodiments, compounds of Formula III may be synthesized by any means known in the art.

[0080] In other non-limiting embodiments, the compounds of Formula III may be synthesized according to the following scheme:

##STR00016## ##STR00017##

wherein R.sup.1, R.sup.2, R.sup.3, R.sup.4, R.sup.5, R.sup.6, R.sup.7, R.sup.8 and R.sup.9 are defined as above for Formula III.

5.3 Methods of Treatment

[0081] In accordance with the invention, there are provided methods of using the compounds of Formulas I-III. The compounds of the instant disclosure can inhibit NFkB activity to exert beneficial effects. A compound of Formula I, II or III that inhibits NF.kappa.B activity may be used, in an effective amount, for the treatment of conditions including, but not limited to, cancer, tumorogenesis, and inflammatory conditions including, but not limited to, type I hypersensitivity, atopy, anaphylaxis, asthma, osteoarthritis, rheumatoid arthritis, septic arthritis, gout, juvenile idiopathic arthritis, still's disease, ankylosing spondylitis, inflammatory bowel disease, Crohn's disease or inflammation associated with vertebral disc herniation. A compound of Formula I, II or III that inhibits BACE 1 activity may be used, in an effective amount, in the treatment of neurodegenerative diseases, such as Alzheimer's disease. In addition, the present invention is directed to the treatment of diseases related to dysfunction of cell proliferation, the immune system and/or inflammation.

5.3.1 Treatment of Disease Related to Dysfunction of Cell Proliferation, the Immune System and/or Inflammation

[0082] In non-limiting embodiments, the present invention provides for methods of treating diseases related to dysfunction of cell proliferation, the immune system and/or inflammation in a subject in need of such treatment by administration of a therapeutic formulation which comprises at least one compound of Formulas I-III.

[0083] In particular embodiments, the formulation may be administered to a subject in need of such treatment in an amount effective to inhibit NFkB activity. Where the formulation is to be administered to a subject in vivo, the formulation may be administered systemically (e.g. by intravenous injection, oral administration, inhalation, subcutaneous, intramuscular, etc.), intraventricularly, intrathecally, or by any other means known in the art. The amount of the formulation to be administered may be determined using methods known in the art, for example, by performing dose response studies in one or more model system, followed by approved clinical testing in humans.

[0084] In one embodiment, the subject or patient has been diagnosed with, or has been identified as having an increased risk of developing a disease associated with dysfunction of cell proliferation, the immune system and/or inflammation.

[0085] In other non-limiting embodiments, the present invention provides for methods of reducing in a subject, the risk of inflammatory damage comprising administering to the subject, an effective amount of a composition according to the invention. An effective amount may be a local concentration or, in a pharmaceutical composition, an amount that, when administered to a subject, results in a therapeutic benefit.

[0086] According to the invention, an effective amount is an amount of at least one compound of Formulas I-III which reduces one or more clinical symptom of one or more of the aforementioned diseases. In one example, an effective amount is an amount of at least one compound of Formulas I-III that reduces the activity of NF.kappa.B, for example, by stabilizing I.kappa.B.alpha. or by inhibiting nuclear translocation of NF.kappa.B.

[0087] In one non-limiting embodiment, the effective amount of at least one compound of Formulas I-III may be determined, for example, via an in vitro assay wherein the effective amount of a compound of Formulas I-III may be correlated with the compound's ability to stabilize I.kappa.B.alpha.. By way of example, and not of limitation, such an assay may comprise a cell-based assay designed to identify compounds that stabilize I.kappa.B.alpha., and thus reduce nuclear translocation of NFkB. The cell-based in vitro IkB.alpha. stabilization assay may utilize a dual reporter system, such as a luciferase reporter system in the cell line OCILy3, a model for primary tumors of the activated B-cell subtype (ABC) of diffuse large B-cell lymphoma (DLBCL) (FIG. 5). The cell line used in the assay may exhibit a high NF.kappa.B activity level, for example, constitutive NFkB activity levels due to high levels of IKK.beta. activity leading to elevated expression of NFkB targeted genes. IKK and proteasome activity may produce high levels of I.kappa.B.alpha. degradation, liberating NF.kappa.B heterodimers to translocate to the nucleus.

[0088] According to the assay, modulators of IkB.alpha. stability may be identified by measuring changes in the level of expression of an IkB.alpha. reporter, for example, an exogenous IkB.alpha.-luciferase fusion reporter construct expressed by an NFkB-insensitive promoter. IkB.alpha. reporter expression may be measured in the presence or absence of a compound of the invention, wherein a greater increase in IkB.alpha. reporter expression by a cell contacted with the compound compared to the IkB.alpha. reporter expression in a cell not contacted with the compound is correlative with the therapeutic efficacy of the compound.

[0089] In one example, a dual luciferase IkB.alpha. stabilization screen in OCI-Ly3 may be used for high throughput screening (HTS). For example, but not by way of limitation, IkB.alpha. fused to a green light-emitting beetle luciferase and a red light-emitting beetle luciferase expressed in a native form can be used to monitor cell uniformity and non-specific effects. Fold-responsiveness may be further increased by having both reporters under the control of inducible promoters, for example, promoters regulated by doxycycline. Upon doxycycline induction of both luciferase reporters, compounds that increased green luminescence with minimal effects on the red luminescence signal may be identified as IkB.alpha. stabilizers, and thus an inhibitor of NF.kappa.B activity.

[0090] In one non-limiting embodiment, the cell based in vitro IkB.alpha. stabilization assay may be the assay depicted in FIG. 5a.

[0091] In one embodiment, an effective amount of a compound of Formulas I-III may be that amount which stabilizes IkB.alpha., and a greater level of IkB.alpha. stabilization at a lower concentration when compared to a control level of IkB.alpha. stabilization, for example, as exhibited by a known IkB.alpha. stabilizing agent, such as MG-132 (FIG. 6) is correlative with the therapeutic efficacy of the compound.

[0092] In one embodiment, the compound may stabilize IkB.alpha. with an efficacy of at least about 10-20%, at least about 20-50%, at least about 50-100%, at least about 100-200%, at least about 200-400%, or at least about 400-800% when compared to the IkB.alpha. stabilization achieved by a known IkB.alpha. stabilizing agent, such as MG-132.

[0093] In one embodiment, an effective amount of a compound of Formulas may be that amount which stabilizes IkB.alpha. with an EC.sub.50 at a concentration ranging from about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, more preferably from about 50 .mu.M to about 0.01 .mu.M, and more preferably from about 20 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein an EC.sub.50 at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0094] In another non-limiting embodiment, the effective amount of at least one compound of Formulas I-III may be determined, for example, via an in vitro assay wherein the effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the nuclear translocation of NF.kappa.B. By way of example, and not of limitation, such an assay may comprise a cell-based assay that utilizes an agent, for example, a cytokine such as TNF.alpha., to stimulate nuclear translocation of endogenous NFkB. Stimulation by the agent may result in proteasome degradation of IkB.alpha. and subsequent translocation of NFkB from the cytoplasm to the nucleus, while in the absence of such a stimulatory agent, NFkB is sequestered in the cytoplasm due to its binding to IkB.alpha..

[0095] In the nuclear-translocation in vitro assay, nuclear translocation of NFkB, for example, the endogenous p65 RelA subunit, may be detected and/or measured following stimulation with the agent through the use of, for example, but not limited to, fluorescent antibody detection and an automated imaging platform. Compounds of the invention may be contacted with cells of the in vitro assay, wherein a reduction in NFkB nuclear transport compared to a cell not contacted with the compound is indicative of the compound's ability to inhibit NF.kappa.B activity. According to the invention, a reduction in nuclear translocation of NF.kappa.B may be correlative with the compound's therapeutic efficacy.

[0096] In one non-limiting embodiment, the cell based nuclear-translocation in vitro assay may be the assay depicted in FIG. 5b.

[0097] In one embodiment, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to inhibit NF.kappa.B nuclear-translocation, wherein a greater level of inhibition at a lower concentration when compared to a control level of inhibition, for example, as exhibited by a known NFiB nuclear-translocation inhibitor, such as BAY 11-7082 (FIG. 6), is indicative of greater therapeutic efficacy of the compound.

[0098] In one embodiment, an effective amount of a compound of Formulas I-III may be that amount which inhibits NF.kappa.B nuclear-translocation with an efficacy of at least about 10-20%, at least about 20-50%, at least about 50-80%, or at least about 80-100% or more when compared to the NF.kappa.B nuclear-translocation inhibition achieved by a known inhibitor, such as BAY 11-7082.

[0099] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which inhibits NF.kappa.B nuclear-translocation by at least 50% when the compound is administered at a concentration ranging from about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, more preferably from about 50 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein inhibition of NF.kappa.B nuclear-translocation at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0100] In another non-limiting embodiment, the effective amount of at least one compound of Formulas I-III may be determined, for example, via an in vitro assay wherein the effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce expression of an NF.kappa.B-dependent reporter construct, for example, a .beta.-lactamase reporter (NF.kappa.B-bla). By way of example, and not of limitation, such an assay may comprise contacting a cell expressing the NF.kappa.B-dependent reporter construct, and monitoring the level of .beta.-lactamase expression, wherein a decrease in expression compared to a cell not contacted with the compound indicates a reduction in NF.kappa.B activity. According to the invention, the reduction in expression of the NF.kappa.B-dependent reporter may be correlative with the compound's therapeutic efficacy.

[0101] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which reduces expression of an NF.kappa.B-bla construct by at least 50% when the compound is administered at a concentration ranging from about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, more preferably from about 50 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein a reduction of NF.kappa.B-bla expression at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0102] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be an amount which achieves a local concentration at the therapeutic site of about 100 .mu.M to about 0.01 .mu.M, preferably from about 50 .mu.M to about 0.01 .mu.M, more preferably from about 20 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay.

5.3.2 Treatment of Neurodegenerative Diseases

[0103] The present invention provides for methods of treating a neurodegenerative disease in a subject in need of such treatment comprising administering, to the subject, a therapeutically effective amount of at least one compound of Formulas I-III. Non-limiting examples of neurodegenerative diseases include Alzheimer's disease, lewy body dementia, inclusion body myositis, and cerebral amyloid angiopathy.

[0104] In particular embodiments, the present invention provides for methods of treating diseases related to metabolism of APP by BACE1 in a subject in need of such treatment by administration of a therapeutic formulation which comprises an effective amount of at least one compound of Formulas I-III. In particular embodiments, the formulation may be administered to a subject in need of such treatment in an amount effective to inhibit BACE1 activity and/or reduce the presence of sAPP.beta. and/or A.beta.. Where the formulation is to be administered to a subject in viva, the formulation may be administered systemically (e.g. by intravenous injection, oral administration, inhalation, etc.), intraventricularly, intrathecally, or by any other means known in the art. The amount of the formulation to be administered may be determined using methods known in the art, for example, by performing dose response studies in one or more model system, followed by approved clinical testing in humans.

[0105] In one embodiment, the subject or patient has been diagnosed with, or has been identified as having an increased risk of developing a neurodegenerative disease, such as Alzheimer's Disease.

[0106] In other non-limiting embodiments, the present invention provides for methods of reducing in a subject, the risk of neural damage comprising administering, to the subject, an effective amount of a composition according to the invention. An effective amount may be a local concentration or, in a pharmaceutical composition, an amount that, when administered to a subject, results in a therapeutic benefit.

[0107] According to the invention, an effective amount is an amount of at least one compound of Formulas I-III which reduces one or more clinical symptom of one or more of the aforementioned diseases. In one example, an effective amount is an amount of at least one compound of Formulas I-III that reduces the production of sAPP.beta. or A.beta. generated by the metabolism of APP by BACE1.

[0108] In one non-limiting embodiment, the effective amount of at least one compound of Formulas I-III may be determined via an in vitro assay, for example, as described in International Patent Application No. PCT/US2007015938 (Publication No. WO 2008008463), which is incorporated in its entirety herein for all purposes, wherein the effective amount may be correlated with the compound's ability to reduce the level of sAPP.beta.. By way of example, and not of limitation, such an assay may comprise a cell-based modified ELISA assay for measuring sAPP.beta., the secreted ectodomain of .beta.-amyloid precursor protein (APP) following .beta.-secretase (BACE1) cleavage. Such an in vitro assay may be used to identify compounds of Formulas I-III that interfere with the first step of sAPP.beta. generation.

[0109] As described in the Examples below, and depicted in FIG. 8, the sAPP.beta. ELISA assay may comprise cells, for example, SY5Y cells, transfected with a BACE1 reporter construct, such as a GFP-tagged BACE 1 (BACE-GFP), and a wild type APP reporter construct, such as a secreted alkaline phosphatase (SEAP)-tagged wild type APP (SEAP-APPwt). BACE 1 cleavage of the reporter-tagged APP (e.g. SEAP-APPwt) may result in secretion into the media of SEAP-tagged sAPP.beta., which may be collected and specifically captured using an sAPP.beta. cleavage site-specific antibody (e.g. s.beta.wt). Following washing, a substrate may be used, for example, the fluorescent alkaline phosphatase substrate 4-methylumbelliferyl phosphate (4-MUP), to detect the captured SEAP-sAPP.beta..

[0110] In one non-limiting example, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the level of sAPP.beta. detected in the in vitro assay compared to a control cell culture that was not contacted with the candidate compound, wherein a reduction of sAPP.beta. compared to the control cell culture correlates with the compound's therapeutic efficacy.

[0111] In another non-limiting example, an effective amount of a compound of Formulas I-III may be that amount which reduces the level of sAPP.beta. detected in the in vitro assay by at least 0.1, or by at least 0.5, or by at least 1, or by at least 1.5, or by at least 2, or by at least 2.5, or by at least 3, or by at least 3.5, or by at least 4, or by at least 4.5, or by at least 5, or by at least 5.5, or by at least 6 or more standard deviations above a control level of sAPP.beta. reduction detected in the in vitro assay when the compound is tested at a concentration of about 2 .mu.M, or about 10 .mu.M, wherein such a reduction of sAPP.beta. correlates with a compound's therapeutic efficacy. In one embodiment, the control level of sAPP.beta. reduction may be the average sAPP.beta. level in control cell cultures that are not contacted with the candidate compound. In other embodiments, the control level may be the average level of sAPP.beta. reduction achieved by a series of compounds tested in the in vitro assay.

[0112] In one preferred non-limiting embodiment, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce sAPP.beta. levels by about 3 standard deviations greater than a control level of sAPP.beta. reduction when the compound is administered at a concentration of 2 .mu.M in the in vitro assay.

[0113] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about 3 standard deviations greater than a control level of sAPP.beta. reduction when the compound is administered at a concentration of 10 .mu.M in the in vitro assay.

[0114] In another example, an effective amount of a compound of Formulas may be that amount which reduces the level of sAPP.beta. by at least 5%, by at least 10%, by at least 20%, by at least 30%, by at least 40%, by at least 50%, by at least 60%, by at least 70%, by at least 80%, by at least 90%, or by 100% when compared to sAPP.beta. level in a control cell culture that was not contacted with the candidate compound, wherein such a reduction of sAPP.beta. correlates with a compound's therapeutic efficacy.

[0115] In another example, an effective amount of a compound of Formulas may be that amount which reduces the level of sAPP.beta. by at least about 50% compared to a control cell culture that was not contacted with the candidate compound, wherein the compound is tested at a concentration ranging from about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein such a reduction of sAPP.beta. at the above-described concentrations is correlative with the compound's therapeutic efficacy.

[0116] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of about 0.5 .mu.M in the in vitro assay.

[0117] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of about 1 .mu.M in the in vitro assay.

[0118] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of about 2 .mu.M in the in vitro assay.

[0119] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of less than 3 .mu.M in the in vitro assay.

[0120] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of about 5 .mu.M in the in vitro assay.

[0121] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of about 8 .mu.M in the in vitro assay.

[0122] In other preferred non-limiting embodiments, an effective amount of a compound of Formulas I-III may be correlated with the compound's ability to reduce the sAPP.beta. level by about at least 50% when the compound is administered at a concentration of about 10 .mu.M in the in vitro assay.

[0123] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to inhibit the enzymatic activity of a BACE1 enzyme. The compound's BACE1 inhibitory effect may be assayed, for example, through use of a BACE1 FRET Assay kit (Invitrogen Corp., Carlsbad, Calif., U.S.A.), wherein the fluorescence resonance energy transfer (FRET)-based assay measures the cleavage by purified recombinant .beta.-secretase of a peptide substrate corresponding to the BACE1 cleavage site of Swedish mutant APP. In such an assay, a greater reduction in fluorescence in the reaction mixture following incubation with a compound of the invention compared to a control cell culture not contacted with the compound is correlative with the compound's therapeutic efficacy.

[0124] In one example, an effective amount of a compound of Formulas I-III may be that amount which inhibits BACE1 enzymatic activity by at least about 50% compared to a control cell culture that was not contacted with the candidate compound. Preferably the compound is tested at a concentration ranging from about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein such an inhibition of BACE1 enzymatic activity at the above-described concentrations is correlative with the compound's therapeutic efficacy.

[0125] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to reduce the level of A.beta..sub.40 in an in vitro assay that measures the level of A.beta..sub.40 produced in a cell line, for example, an SY5Y-BACEGFP-SEAPAPPwt cell line. In one non-limiting example, the level of A.beta..sub.40 expressed by the cell line may be measured through the use of an A.beta..sub.40 Elisa kit (BioSource). In one embodiment, the assay comprises incubating the A.beta..sub.40 expressing cells with a compound of Formulas I-III, followed by assaying the concentration of A.beta..sub.40 in the cell culture medium. In such an assay, a greater reduction of A.beta..sub.40 concentration in the cell culture medium following incubation with a compound compared to a control cell culture not contacted with the compound is correlative with the compound's therapeutic efficacy.

[0126] In one example, an effective amount of a compound of Formulas I-III may be that amount which reduces the level of A.beta..sub.40 in an in vitro assay by at least about 50% compared to a control cell culture that was not contacted with the candidate compound. Preferably the compound is tested at a concentration ranging from about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein such a reduction in the level of A.beta..sub.40 at the above-described concentrations is correlative with the compound's therapeutic efficacy.

[0127] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to reduce the level of A.beta..sub.40 in an in vitro assay that measures the level of A.beta..sub.40 produced in a cell culture, for example, a culture of primary cortical neurons transduced with lentivirus carrying Swedish mutant APP (APPsw). In one embodiment, the assay comprises incubating the A.beta..sub.40 expressing cells with a compound of Formulas I-III, followed by assaying the concentration of A.beta..sub.40 in the cell culture medium. In such an assay, a greater reduction of A.beta..sub.40 concentration in the cell culture medium following incubation with a compound compared to a control cell culture not contacted with the compound is correlative with the compound's therapeutic efficacy.

[0128] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which reduces A.beta..sub.40 by about 1-10%, more preferably from about 10-20%, more preferably from about 20-30%, more preferably from about 30-40%, more preferably from about 40-50%, more preferably from about 50-60%, more preferably from about 60-70%, more preferably from about 70-80%, more preferably from about 80-90%, and more preferably from about 90-100%, compared to A.beta..sub.40 levels in the cell culture medium of a control cell culture that was not incubated with the compound. Preferably the compound is incubated at a concentration of about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein a greater level of A.beta..sub.40 reduction at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0129] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to reduce the level of sAPP.beta. in an in vitro assay that measures the level of sAPP.beta. produced in a cell culture, for example, primary cortical neurons transduced with lentivirus carrying Swedish mutant APP (APPsw). In one embodiment, the assay comprises incubating the cells with a compound of Formulas I-III, followed by assaying the concentration of sAPP.beta. in the cell culture medium. In such an assay, a greater reduction of sAPP.beta. concentration in the cell culture medium following incubation with a compound compared to a control cell culture not contacted with the compound is correlative with the compound's therapeutic efficacy.

[0130] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which reduces sAPP.beta. by about 1-10%, more preferably from about 10-20%, more preferably from about 20-30%, more preferably from about 30-40%, more preferably from about 40-50%, more preferably from about 50-60%, more preferably from about 60-70%, more preferably from about 70-80%, more preferably from about 80-90%, and more preferably from about 90-100%, compared to sAPP.beta. levels in the cell culture medium of a control cell culture that was not incubated with the compound. Preferably the compound is incubated at a concentration of about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein a greater level of sAPP.beta. reduction at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0131] In other non-limiting embodiments, an effective amount of a compound of Formulas I-III may be that amount which reduces A.beta..sub.40 by about 50% in an in vitro assay compared to A.beta..sub.40 levels in the cell culture medium of a control cell culture that was not incubated with the compound. Preferably the compound is incubated at a concentration of about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to 0.01 .mu.M in the in vitro assay, wherein a reduction of A.beta..sub.40 at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0132] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to reduce the level of A.beta..sub.40 in an in vitro assay that measures the level of A.beta..sub.40 produced in a cell culture, for example, cultured primary cortical neurons prepared from Tg2576 mice (i.e. mice carrying human APPsw transgene under the control of the PrP promoter). In one embodiment, the assay comprises incubating the A.beta..sub.40 expressing cells with a compound of Formulas I-III, followed by assaying the concentration of A.beta..sub.40 in the cell culture medium. In such an assay, a greater reduction of A.beta..sub.40 concentration in the cell culture medium following incubation with a compound compared to a control cell culture not contacted with the compound is correlative with the compound's therapeutic efficacy.

[0133] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which reduces A.beta..sub.40 by about 1-10%, more preferably from about 10-20%, more preferably from about 20-30%, more preferably from about 30-40%, more preferably from about 40-50%, more preferably from about 50-60%, more preferably from about 60-70%, more preferably from about 70-80%, more preferably from about 80-90%, and more preferably from about 90-100%, compared to A.beta..sub.40 levels in the cell culture medium of a control cell culture that was not incubated with the compound. Preferably when the compound is incubated at a concentration of about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay, wherein a greater level of A.beta..sub.40 reduction at a lower concentration in the in vitro assay is correlative with the compound's therapeutic efficacy.

[0134] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to reduce the level of A.beta..sub.40 and/or sAPP.beta. in an ex vivo assay that measures the level of A.beta..sub.40 and/or sAPP.beta. produced in cells, for example, organotypic brain slices from Tg2576 mice (i.e. mice carrying human APPsw transgene under the control of the PrP promoter). In one embodiment, the assay comprises incubating the brain slices with a compound of Formulas followed by assaying the concentration of A.beta..sub.40 and/or sAPP.beta. in the brain slices. In such an assay, a greater reduction of A.beta..sub.40 and/or sAPP.beta. concentration in the brain slices following incubation with a compound compared to a control brain slice not contacted with the compound is correlative with the compound's therapeutic efficacy.

[0135] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which reduces A.beta..sub.40 and/or sAPP.beta. by about 1-10%, more preferably from about 10-20%, more preferably from about 20-30%, more preferably from about 30-40%, more preferably from about 40-50%, more preferably from about 50-60%, more preferably from about 60-70%, more preferably from about 70-80%, more preferably from about 80-90%, and more preferably from about 90-100%, compared to A.beta..sub.40 and/or sAPP.beta. levels in control brain slices that were not incubated with the compound. Preferably the compound is incubated at a concentration of about 200 .mu.M to about 0.01 .mu.M, preferably from about 100 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the ex vivo assay, wherein a greater level of A.beta..sub.40 and/or sAPP.beta. reduction at a lower concentration in the ex vivo assay is correlative with the compound's therapeutic efficacy.

[0136] In other non-limiting embodiments, the effective amount of at least one compound of Formulas I-III may be correlated with the compound's ability to reduce the level of A.beta..sub.40 and/or sAPP.beta. in an in vivo assay that measures the level of A.beta..sub.40 and/or sAPP.beta. produced in a test subject, for example, a Tg2576 mouse (i.e. mice carrying human APPsw transgene under the control of the PrP promoter). In one embodiment, the assay comprises administering at least one compound of Formulas I-Ill to the test subject, for example, via interstitial fluid (ISF) compound administration, intraperitoneal (IP) compound injection, or through the use of a microdialysis apparatus for infusion of the compound at multiple concentrations (for example, in the hippocampus of the test subject), followed by assaying the concentration of A.beta..sub.40 and/or sAPP.beta. in the test subject. In such an assay, a greater reduction of A.beta..sub.40 and/or sAPP.beta. concentration in the test subject following administration of the compound compared to a control subject not administered the compound is correlative with the compound's therapeutic efficacy.

[0137] In one non-limiting embodiment, an effective amount of a compound of Formulas I-III may be that amount which reduces A.beta..sub.40 and/or sAPP.beta. by about 1-10%, more preferably from about 10-20%, more preferably from about 20-30%, more preferably from about 30-40%, more preferably from about 40-50%, more preferably from about 50-60%, more preferably from about 60-70%, more preferably from about 70-80%, more preferably from about 80-90%, and more preferably from about 90-100%, compared to A.beta..sub.40 and/or sAPP.beta. levels in brain homogenates of subjects that were not administered the compound. Preferably the compound is administered at a concentration of about 0.5 mg/kg to about 20 mg/kg, preferably from about 1 mg/kg to about 20 mg/kg, more preferably from about 3 mg/kg to about 20 mg/kg, more preferably from about 5 mg/kg to about 20 mg/kg, more preferably from about 10 mg/kg to about 20 mg/kg in the in vivo assay, wherein a greater level of A.beta..sub.40 and/or sAPP.beta. reduction at a lower concentration in the in vivo assay is correlative with the compound's therapeutic efficacy.

[0138] In non-limiting embodiments, an effective amount of a compound of Formulas I-III may be an amount which achieves a local concentration at the therapeutic site of about 100 .mu.M to about 0.01 .mu.M, preferably from about 50 .mu.M to about 0.01 .mu.M, more preferably from about 20 .mu.M to about 0.01 .mu.M, and more preferably from about 10 .mu.M to about 0.01 .mu.M in the in vitro assay.

5.3.3 Administration of Treatments

[0139] According to the invention, the component or components of a pharmaceutical composition of the invention may be administered to a subject by means including but not limited to intravenous, intra-arterial, intramuscular, intradermal, transdermal, subcutaneous, oral, intraperitoneal, intraventricular, and intrathecal administration.

[0140] In particular non-limiting embodiments, the therapeutic compound can be delivered in a controlled or sustained release system. For example, a compound or composition may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In one embodiment, a pump may be used (see Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574). In another embodiment, polymeric materials can be used (see Langer and Wise eds., 1974, Medical Applications of Controlled Release, CRC Press: Boca Raton, Fla.; Smolen and Ball eds., 1984, Controlled Drug Bioavailability, Drug Product Design and Performance, Wiley, N.Y.; Ranger and Peppas, 1983, J. Macromol. Sci. Rev. Macromol. Chem., 23:61; Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neural., 25:351; Howard et al., 9189, J. Neurosurg. 71:105). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., the heart or a blood vessel, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, 1984, in Medical Applications of Controlled Release, supra, Vol. 2, pp. 115-138). Other controlled release systems known in the art may also be used.

5.4 Pharmaceutical Compositions

[0141] The compounds and compositions of the invention may be formulated as pharmaceutical compositions by admixture with a pharmaceutically acceptable carrier or excipient.

[0142] For example, the pharmaceutical composition may comprise an effective amount of at least one compound of Formulas I-III and a physiologically acceptable diluent or carrier. The pharmaceutical composition may further comprise a second drug, for example, but not by way of limitation, an anti-cancer drug, an anti-inflammatory drug, for example, but not limited to, a steroid compound and/or a non-steroidal anti-inflammatory drug, or compound for the treatment of Alzheimer's disease, such as an acetylcholinesterase inhibitor or an NMDA glutamate receptor antagonist (e.g. memantine).

[0143] The phrase "pharmaceutically acceptable" indicates that a substance is physiologically tolerable when administered to a subject. Preferably, but not by way of limitation, as used herein, the term "pharmaceutically acceptable" means approved by a regulatory agency of the federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, or, for solid dosage forms, may be standard tabletting excipients. Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin, 18th Edition, or other editions.

[0144] In a specific embodiment, the therapeutic compound can be delivered in a vesicle, in particular a liposome (see Langer, 1990, Science 249:1527-1533; Treat et al., 1989, in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler eds., Liss: New York, pp. 353-365; Lopez-Berestein, ibid., pp. 317-327; see generally Lopez-Berestein, ibid.).

[0145] The present invention is not to be limited in scope by the specific embodiments described herein and the Examples that follow. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying Examples and Figures. Such modifications are intended to fall within the scope of the appended claims.

[0146] Patents, patent applications, publications, product descriptions, GenBank Accession Numbers, and protocols are cited throughout this application, the disclosures of which are incorporated herein by reference in their entireties for all purpose.

EXAMPLES

Example 1

Synthesis of Compounds of Formulas I-III

[0147] The compounds of Formulas I and II consist of N-(quinolin-8-yl)benzensulfonamides, and the compounds of Formula III consist of a structurally constrained cyclic sulfonamide with the sultam bridging quinolinyl and phenyl moieties. This particular scaffold lacks a general synthetic method. Described here is an efficient synthesis of this class of compounds that allows a diverse substitution pattern.

[0148] The synthetic strategy was to generate N-8-quinolinyl benzenesulfonamides, and then construct the sultam skeleton through cyclization of the easily accessible N-8-quinolinyl benzenesulfonamides. Diazotization-induced cyclization was employed to synthesize the target compound (FIG. 2). Sulfonamide 8 was prepared from 2-nitrobezenesufonyl chloride and 8-aminoquinoline 2 in pyridine. The nitro group was reduced with SnCl.sub.2 to provide amine 9 and subsequent diazotization afforded triazine 10. A procedure employing Cu (0) and NaOH (Ullman et al., 1911, Ber. Dtsch. Chem. Ges. 43:2694), failed to yield the desired product, but thermolysis in a variety of solvents (e.g. EtOH, H.sub.2O, DMSO or HOAc) or neat afforded the product 1. The best result (41% yield) was obtained by heating in glacial acetic acid at 120.degree. C. for ten minutes.

[0149] A second synthetic method involves a one-pot synthesis of N-8-quinolinyl benzenesultams from N-8-quinolinyl-2-aminobezenesulfonamides. In a typical reaction, as shown in FIG. 3A, 2-aminobenzenesulfonamide 9a, prepared from 6-methoxy-2-methylquinolin-8-amine 2a (Qiu et al., 2007, Inorg. Chim. Acta. 360:431), was dissolved in HOAc and treated with 1.5 equiv of t-BuONO at 10.degree. C. The reaction was allowed to warm to room temperature over 10 minutes and afforded sultam 1a in (78% yield) without isolation of the triazine intermediate.

[0150] A selection of N-8-quinolinyl-2-aminobezenesulfonamides (9a-p) were prepared and subsequently cyclized in similar one-pot reactions to give sultams (1a-p) (FIG. 4). In each case, the product was isolated without intermediate purification (FIG. 4). Aminobezenesulfonamides (9b-h and 9l-p) were synthesized in a similar manner described in FIG. 3A. Intermediates 9i-k were prepared from aniline 11 as outlined in FIG. 3B: Sulfonamidation of aniline 11 with 2-nitrobezenesufonyl chloride provided sulfonamide 12. Skraup reaction under modified conditions (Matsugi et al., 2000, Tetrahedron Lett. 41:8523) followed by reduction of nitro group provided the N-quinolinyl sulfonamides (9i-k).

[0151] A typical synthesis of a compound of Formulas II and III involves the following steps:

[0152] Compound 9a: To a solution of 6-methoxy-2-methylquinolin-8-amine 2a (0.19 g, 1.0 mmol) in pyridine (5 ml) was added 4-methyl-2-nitrobenzenesulfonyl chloride (0.24 g, 1.0 mmol). The mixture was stirred at room temperature overnight and precipitated with H.sub.2O. The crude product was filtered and recrystallized from EtOH to afford nitrobenzenesulfonamide (0.31 g, 79%) as red crystal. .sup.1H NMR (300 MHz, CDCl.sub.3) .delta. 10.1 (br, 1H); 8.04 (d, 1H), 7.86 (d, 1H), 7.62 (m, 2H), 7.39 (d, 1H), 7.24 (d, 1H), 6.73 (s, 1H), 3.88 (s, 31-1), 2.66 (s, 3H), 2.41 (s, 3H); ESI-MS (M.sup..+-..+-.1): 388.0. To a suspension of above nitro compound (0.20 g, 0.56 mmol) in EtOH (5 ml) was added SnCl.sub.2 (0.32 g, 1.7 mmol) slowly. The mixture was refluxed for 2 h. After removal of EtOH, the residue was treated with 1M NaOH. The aqueous solution was extracted with CH.sub.2Cl.sub.2. The combined organic phases were washed by brine, dried over Na.sub.2SO.sub.4, and concentrated to yield 9a (0.18 g, 92%) as white solid. .sup.1H NMR (300 MH.sub.z, CDCl.sub.3) .delta. 9.42 (br, 1H); 7.83 (d, 1H), 7.65 (d, I H), 7.30 (s, 1H), 7.23 (d, 1H), 6.64 (s, 1H), 6.48 (d, 1H), 6.42 (s, 111), 3.80 (s, 3H), 2.61 (s, 3H), 2.17 (s, 3H); ESI-MS (M.sup.++1): 358.1.

[0153] Compound 1a:

[0154] To a solution of 9a (0.1 g, 0.28 mmol) in HOAc (1 ml) at 10.degree. C. was added t-BuONO (0.05 mL, 0.42 mmol). The reaction was slowly warmed to room temperature over 10 min and quenched with H.sub.2O. The mixture was extracted with EtOAc. The combined organic phases were washed with saturated NaHCO.sub.3, dried over Na.sub.2SO.sub.4, and concentrated. Flash chromatography (EtOAc/CH.sub.2Cl.sub.2 1:10 vv) gave 1a (74 mg, 78%) as yellow solid. .sup.1H NMR (300 MHz, CDCl.sub.3) .delta. 8.49 (s, 1H), 7.98 (d, 1H), 7.92 (d, 1H), 7.37 (d, 114), 7.25 (d, 1H), 6.87 (s, 1H), 4.00 (s, 3H), 2.69 (s, 3H), 2.52 (s, 3H); .sup.13C NMR (75 MHz, CDCl.sub.3) .delta. 156.2, 155.2, 142.2, 134.8, 134.5, 134.1, 132.2, 131.4, 130.1, 129.0, 126.8, 124.4, 122.0, 112.5, 100.0, 56.2, 25.2, 22.6; ESI-MS (M.sup.++1): 341.2.

[0155] In summary, a one-pot reaction for the conversion of N-8-quinolinyl-2-aminobezenesulfonamides into their corresponding sultams under mild conditions has been described. This procedure offers access to a class of heterocyclic compounds that have shown therapeutic potential as novel BACE1 and NF-.kappa.B inhibitors, as described below.

Example 2

Inhibition of NFkB Activity

[0156] Small molecule regulation of signaling cascades associated with NFkB may provide novel approaches to alleviate numerous disease states, (D'Acquisto, et al., 2006 Curr. Opin. Pharmacol. 6:387; Gilmore, et al., 2006, Oncogene 25:6887). Several high-throughput campaigns in search of novel inhibitors of the NFkB signal transduction pathway have been undertaken. Two of these include a cell-based assay designed to identify IkB.alpha. stabilization, and a cell-based assay designed to identify inhibitors of TNF.alpha.-induced translocation of NFkB. Despite the differing assay design, cell type, and signal readout, a N-(quinolin-8-yl)benzenesulfonamide core scaffold was identified in both screens.

NFkB Inhibitory Assays

[0157] The first assay, an IkB.alpha. stabilization screen, was performed using a dual luciferase reporter system in the cell line OCILy3, an excellent model for primary tumors of the activated B-cell subtype (ABC) of diffuse large B-cell lymphoma (DLBCL) (Figure SA; PubChem AID: 445) (Davis, et al., 2007, Drug Dev. Technol. 5:85; Davis, et al., 2001, J. Exp. Med. 184:1861; Okamoto, et al., 2007, Curr. Pharm. Des. 13:447) Abnormally high constitutive NFkB activity levels have been noted in ABC-DLBCL, as in several types of cancer, due to high levels of IKK13 activity leading to elevated expression of NFkB targeted genes. (Okamoto, et al., 2007, Curr. Pharm. Des. 13:447) In this setting, IKK and proteasome activity produce high degradation of IkB.alpha., liberating p50/65 and/or p50/c-Rel heterodimers to translocate to the nucleus. Because lines of ABC-DLBCL and other cancer types are dependent on constitutive NFkB activity, the NFkB pathway is a therapeutic target. This is particularly true for those processes governing IkB.alpha. degradation, and the therapeutic potential of small molecules for this purpose has been shown by studies in ABC-DLBCL lines using a specific IKK.beta. inhibitor. Furthermore, the use of an ABC-DLBCL line for a small-molecule screen of IkB.alpha. stabilization provides a context that is especially close to that of the targeted disease, with the potential that inhibitors may be found that affect specific upstream points in IKK activation.

[0158] To identify modulators of IkB.alpha. stability in ABC-DLBCL lines, such as small molecules inhibiting IKK or proteasome activity, changes in the level of an exogenous IkB.alpha.-luciferase fusion reporter expressed by an NFkB-insensitive promoter were measured. (Lam et al., 2005, Clin. Cancer res. 11:28; Ngo et al., 2006, Nature 441:106) The dual luciferase IkB.alpha. stabilization screen in OCI-Ly3 employed was designed to be suitable for HTS, by using IkB.alpha. fused to a green light-emitting beetle luciferase, with a red light-emitting beetle luciferase expressed in a native form to monitor cell uniformity and non-specific effects. (Davis, et al., 2007, Drug Dev. Technol. 5:85) Fold-responsiveness was further increased by having both reporters under the control of inducible promoters regulated by doxycycline. Upon doxycycline induction of both luciferase reporters, compounds that increased green luminescence with minimal effects on the red luminescence signal were scored as IkB.alpha. stabilizers. (Davis, et al., 2007, Drug Dev. Technol. 5:85).

[0159] The second assay, a translocation-based assay, was a high-content screen performed in human umbilical vein endothelial cells (HUVEC) using TNF.alpha. to stimulate nuclear translocation of endogenous NFkB (Figure SB; PubChem AID: 438). NFkB is sequestered in the cytoplasm due to its binding to IkB.alpha., which blocks exposure of a nuclear localization sequence. Activation by cytokines such as TNF.alpha. results in proteasome degradation of IkB.alpha. and subsequent translocation of NFkB from the cytoplasm to the nucleus. In the assay, nuclear translocation of the endogenous p65 RelA subunit of NFkB, at 30 min post-stimulation, was monitored using fluorescent antibody detection and an automated imaging platform. (Mayer et al., 2006, Methods Enzyme 414:266) NFkB inhibitors in this assay, such as IkB.alpha. stabilizers, were detected as compounds that interfered with p65 translocation to the nucleus.

[0160] The above-described assays were validated using known inhibitors of the NFkB pathway (FIG. 6). The proteasome inhibitor MG-132 (1) 16 served as a positive control for the IkB.alpha. stabilization assay. The translocation assay used BAY 11-7082 (2), an agent that inhibits the TNF.alpha.-induced phosphorylation of IkB.alpha.. (Kamthong, 2001, Biochem 356:525; Izban et al., 2000, Hum. Pathol. 31:1482; Pierce et al., 1997, J. Biol. Chem. 272:21096). Additionally, a substituted 2-(thiophen-2-yl)quinazoline 3 which acts as an inhibitor of NFkB and AP1 mediated transcriptional activation (Palanki et al., 2003, Bioorg. Med. Chem. Lett. 13:4077) was used as a positive control in a secondary assay that used TNF.alpha. to stimulate NFkB-dependent expression of a .beta.-lactamase reporter (NFkB-bla in FIG. 7). This compound also served as a negative control for the dual luciferase IkB.alpha. stabilization assay.

Results

[0161] N-(quinolin-8-yl)benzenesulfonamide (4) and the related C7-locked N-(quinolin-8-yl)benzenesulfonamide (5) were identified as two common structures in both screens (FIG. 6). To further confirm that these agents interfered with the NFkB activation in a genuine manner, a reporter assay obtained from Invitrogen where induction of .beta.-lactamase occurred in an NFkB-dependent manner was also performed on the active compounds. These data confirmed the ability of these compounds to inhibit the NFkB pathway, and the compounds were then advanced for further study (FIG. 7).

[0162] The synthesis of analogues of (4) was undertaken through a one-step process involving the addition of unsubstituted or 2-substituted, 6-substituted or 2,6-disubstituted quinolin-8-amines and a variety of sulfonyl chlorides (FIG. 7B). Amino-substituted analogues (19-22) (Izban et al., 2000, Hum. Pathol. 31:1482) were obtained by reduction of the corresponding nitro compounds with stannous chloride in ethanol (FIG. 7D). Over 40 novel analogues were prepared to establish preliminary structure activity relationships.

Discussion

[0163] The synthesis of various analogues of (5) via a diazotization-induced cyclization of accessible N-8-quinolinyl-2-aminobenzenesulfonamides proved to be an efficient way of synthesizing this type of C7-locked sulfonamide (FIG. 7D).

[0164] Analogues of (4) and (5) were analyzed within both screens. Results for analogues of (4) are shown in FIG. 7A, while the results for derivatives of (5) are shown in FIG. 7C. In general, there was agreement in the relative potencies of analogues tested in both screening assays, as well as the NFkB .beta.-lactamase reporter assay (although exceptions do exist).

[0165] A more rigid structure was associated with compound 5, because a bridge between the quinoline carbon 7 and the ortho-position of the sulfonyl phenyl ring locks the core scaffold in a rigid conformation. Two compounds (30 and 31) with the best overall potency values were identified here. Substituents at the 9 position (para to the sulfonylamide moiety designated R''' in FIG. 7C) are generally well tolerated as are 6-methoxy and 2-methyl substitutions (analogues 32 and 37-40, respectively).

Example 3

High- and Medium-Throughput Screening of Small Molecule Libraries to Identify Small Molecule Modulators of BACE1

[0166] A cell-based modified ELISA assay for measuring sAPP.beta., the secreted ectodomain of .beta.-amyloid precursor protein (APP) following .beta.-secretase (BACE 1) cleavage, was used to identify a class of compounds that interfered with the first step of sAPP.beta. generation. This assay has been described in International Application PCT/US2007015938 (Published as International Publication No. WO 08/008463), which is herein incorporated in its entirety for all purposes.

[0167] BACE1-mediated cleavage of APP is a key and necessary event in the generation of neurotoxic .beta.-amyloid (A.beta.), a widely accepted contributor to the development of Alzheimer's disease (AD). Studies in BACE 1 knockout mice showed that they are viable, fertile, and do not produce A.beta., making BACE1 an attractive target for AD therapeutic intervention.

[0168] The SY5Y-BACEGFP-SEAPAPPwt cell based assay was developed to discover novel small molecule modulators of BACE1 activity. SY5Y cells were stably transfected with GFP-tagged BACE1 (BACE-GFP) and secreted alkaline phosphatase (SEAP)-tagged wildtype APP (SEAP-APPwt). BACE1 cleavage of SEAP-APPwt results in secretion into the media of SEAP-tagged sAPP.beta., which is collected and specifically captured using an sAPP.beta. cleavage site-specific antibody (s.beta.wt). After washing, the fluorescent alkaline phosphatase substrate 4-methylumbelliferyl phosphate (4-MUP) is used to detect the captured SEAP-sAPP.beta. (FIG. 8).

Targeted Screening Using N-(Quinolin-8-Yl)Benzenesulfonamides and Related Compounds

[0169] N-(quinolin-8-yl)benzenesulfonamides (FIG. 2) were recently reported as agents capable of down-regulating NF-.kappa.B activity (Xie et al., 2008). NF-.kappa.B is a ubiquitous transcription factor that is thought to play a role in many cellular processes, including immune and inflammatory responses, developmental processes, cellular growth, and apoptosis. Dysregulation of NF-.kappa.B signaling has been associated with many disease states, including asthma, arthritis, cancer, inflammatory conditions, neurodegenerative diseases, and heart disease (reviewed in Kumar et al., 2004). NF-.kappa.B inhibitors have also been reported to inhibit Ap production (Paris et al., 2007). Furthermore, N-(quinolin-8-yl)benzenesulfonamides bear structural similarity to clioquinol and its second-generation compound PBT2. Both clioquinol and PBT2 are believed to be strong metal ion chelators that may interfere with oligomerization and aggregation of AP and promote its clearance (Ritchie et al., 2003). PBT2, developed by Prana Biotechnology, is currently in Phase II clinical trial for the treatment of AD. Thus, a small library of N-(quinolin-8-yl)benzenesulfonamides and related compounds was screened for agents that reduce BACE1-mediated cleavage of APP.

[0170] 176 compounds were tested in the primary screen (FIG. 9). Briefly, each compound (10 mM stock in DMSO) was diluted 1:25 in DMSO and loaded onto 96-well plates to generate a 400 .mu.M stock plate. 1 .mu.l of each compound was then diluted 1:200 with cell culture media (0.5% DMSO concentration) in two separate 96-well plates for a final screening concentration of 2 .mu.M. 150 .mu.l of media containing compound was then transferred onto SY5Y-BACEGFP-SEAPAPPwt cells in 96-well format, and the BACE1 assay was performed as described previously. The primary screen was conducted in duplicate. A parallel screen was conducted at 10 .mu.M concentration, also in duplicate. This data is not shown. Percent inhibition of the fluorescence signal was calculated as 100.times.[DMSO control compound]/DMSO control The majority of data points were clustered between -25% and 25% inhibition of sAPP.beta. fluorescence signal (FIG. 9A). The Z' factors for all 4 96-well plates used in the primary screen were above 0.7 (data not shown). The threshold for hit selection was set at 3 standard deviations, or 59.76% inhibition, yielding 6 small molecule hits and a hit rate of 3.41% (FIG. 9B).

[0171] Several partial hits were also identified in the targeted screen of N-(quinolin-8-yl)benzenesulfonamides. Partial hits were defined loosely as compounds that either exhibited activity just below the threshold 59.76% inhibition in the 2 .mu.M screen or those that displayed significant activity in the 10 .mu.M screen. These compounds were characterized for 8-point dose-response and structure-activity relationship studies.

[0172] In summary, a cell-based BACE1 assay was used to screen N-(quinolin-8-yl)benzene-sulfonamides and related compounds, a class of NF-.kappa.B inhibitors. Because of their potency, availability, and the reported connection between NF-.kappa.B inhibition and AP generation, structural analogs of N-(quinolin-8-yl)benzenesulfonamides were also selected for investigation in structure-activity relationship studies. Primary and secondary screening using the cell-based BACE1 assay thus yielded chemotypes for SAR studies and further characterization in more physiological systems.

Example 4

Structure-Activity Relationship Studies and Characterization in Physiological Systems

[0173] SAR studies were conducted for full and partial hits from the targeted screen of N-(quinolin-8-yl)benzenesulfonamides. Characterization in SY5 Y-BACEGFP-SEAPAPPwt cells identified CU-264 as the most potent analog. CU-264 was further characterized in a of more physiological assay for its ability to reduce A.beta..sub.40 and sAPP.beta.. CU-264 had the effect of raising A.beta..sub.40 and sAPP.beta. levels.

SAR Studies of N-(quinolin-8-yl)Benzenesulfonamides

[0174] Targeted screening of N-(quinolin-8-yl)benzenesulfonamides and related compounds revealed several full and partial hits capable of reducing the fluorescent signal from the cell-based BACE1 assay. Based on the chemical structures of the 176 compounds as well as the full and partial hits from the targeted screen, the structural elements that were critical for activity were identified. All full and partial hits contained the sulfonamide of quinoline motif either in open or cyclized varieties, as shown in FIG. 10. Substituting naphthalene for quinoline or removal of the sulfonamide substituent resulted in loss of activity.

[0175] 28 full and partial hits were characterized in 8-point dose-response experiments for IC.sub.50 determination in SY5Y-BACEGFP-SEAPAPPwt cells (FIG. 10). The investigation revealed 3 compounds with sub-micromolar potencies for sAPP3 reduction--CU-163, CU-264, and CU-262. Analysis of the data revealed a few structure-activity relationships. Addition of a methyl group at R3 resulted in a reduction of potency (compare CU-274, CU-278, CU-280, and CU-281 with CU-242, CU-271, CU-265, and CU-264, respectively). The electronic properties of the R groups did not affect compound activity based on the available data. The addition of electron-withdrawing, electron-donating, and electron-neutral groups did not have a clear and consistent effect on activity. Although there are no direct comparisons, cyclization of compounds (e.g. CU-262 and CU-282) did not significantly alter the potency compared with non-cyclized compounds (e.g. CU-264, CU-265, CU-280, and CU-281).

[0176] SAR studies of N-(quinolin-8-yl)benzenesulfonamides revealed 3 compounds with sub-micromolar potencies for sAPP.beta. reduction. The most potent compound, CU-264, is further evaluated.

Evaluation of CU-264 in More Physiological Systems

[0177] The previously described hit was further analyzed in physiologically relevant systems, e.g. primary neurons and Alzheimer's model mice. Two complementary neuronal cell systems were employed to test the effects of candidate compounds on various aspects of APP processing: cultured mouse cortical neurons (postnatal day 0) infected with recombinant lentivirus carrying human APPsw (Lenti-APPsw; FIG. 11); and cultured cortical neurons prepared from Tg2576 mice (carrying human APPsw transgene under the control of the PrP promoter). Lenti-APPsw infected cortical neurons can be prepared in a relatively large scale, but suffer from variability in lentiviral infection efficiency, resulting in variable levels of APP expression. In contrast, cortical neurons derived from Tg2576 provide constant APP expression. However, neuronal yield is generally much lower, since only half the pups will contain the transgene when heterozygote crossbreeds were conducted. Therefore, Initial characterization experiments were performed using cortical neurons infected with Lenti-APPsw. Compounds exhibiting inhibitory activity on sAPP.beta. and/or A.beta. would then be re-evaluated in Tg2576 mice-derived neurons.

[0178] Ex vivo and in vivo assays will be used to characterize certain hit(s). Ex vivo systems, such as organotypic brain slices, offer an alternative to in vivo assays, and can also be used to test a large number of compounds. Further more, at least with regard to the A.beta. release phenotype, brain slice data have been shown to correlate well with the results obtained in vivo (reviewed in Noraberg et al., 2005). Brain slices from p7 Tg2576 pups are used herein for compound characterization.

[0179] For in vivo studies, interstitial fluid (ISF) compound administration and A.beta. measurement, as well as intraperitoneal (IP) compound injection, both utilizing Tg2576 mice, will be used to characterize compounds. Positioning of a guide cannula to the mouse hippocampus allows for insertion of a microdialysis apparatus, which can be used to infuse compounds at multiple concentrations sequentially in the awake mouse. A.beta. measurements can be performed using the same apparatus, yielding rapid dose-response determinations (Cirrito et al., 2003). This method allows for rapid assessment of compound effect on AP on a dynamic time scale. More conventionally, compounds can also be administered via IP injection. For these experiments, 12-month old Tg2576 mice will be used.

CU-264 does not Reduce A.beta.40 in Lenti-APPsw Infected Primary Cortical Neurons

[0180] Lenti-APPsw infected primary cortical neurons were used for the initial round of physiological experiments. Primary cortical neurons were harvested from wild-type P0 pups using established protocols. The majority of cells from the resulting culture exhibit neuronal morphology on light microscopy and express neuronal .beta.-tubulin as visualized by immunocytochemistry using the TUJ1 antibody (Covance, FIG. 12).

[0181] The Lenti-APPsw vector was co-transfected into HEK293 T cells with ViraPower packaging mix (Invitrogen) to generate the lentivirus. Lentiviral-mediated transduction of APPsw in primary neurons was performed by adding neuron primary culture media containing the lentiviral particles to wild-type DIV-14 primary neurons (FIG. 11). After 24 hours, neurons were incubated with media for 72 hours prior to collecting for A.beta..sub.40 measurement. Two batches of virus (LV-1 and LV-2) were tested, showing that there is batch-to-batch variation in the viral titer.

[0182] Using this experimental paradigm, CU-264 was tested at 10 and 2 .mu.M concentrations to examine the sAPP.beta.- and A.beta.-lowering activity of the compound. The protocol was modified slightly to allow for 24-hour compound treatment. DIV-14 wild-type primary neurons (FIG. 12) cultured on 6-well plates were incubated for 24 hours with primary culture media containing lentiviral particles. After infection, fresh media was mixed in a 1:1 ratio with conditioned media collected prior to lentiviral infection, and applied to the cells for 48 hours to allow for APP expression. At the end of this "pre-drug" incubation, media containing compound was applied for 24-hour treatment. Control experiments using DMSO alone showed that media collected after the 24-hour treatment period contained at least two times higher A.beta..sub.40 than that collected just after the 48-hour pre-drug period. CU-264, which exhibited sub-micromolar potency for sAPP.beta. reduction in SY5Y-BACEGFP-SEAPAPPwt cells, did not reduce A.beta..sub.40 in primary neurons (FIG. 13A). In contrast, CU-264 was observed to increase AP at 10 .mu.M.

[0183] The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims.

[0184] Patents, patent applications, publications, product descriptions, GenBank Accession Numbers, and protocols are cited throughout this application, the disclosures of which are incorporated herein by reference in their entireties for all purposes.

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed