Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
RAPAMYCIN ANALOG FOR PREVENTION AND/OR TREATMENT OF NEURODEGENERATIVE CONDITIONS
Document Type and Number:
WIPO Patent Application WO/2019/157382
Kind Code:
A1
Abstract:
This disclosure relates to a novel rapamycin analogue of Formula I or Formula II, mixtures, methods for its production, and its use in preventing and/or treating a neurodegenerative condition.

Inventors:
POWERS TIMOTHY S (US)
COX BRYAN F (US)
MARSHAK RICHARD B (US)
Application Number:
PCT/US2019/017355
Publication Date:
August 15, 2019
Filing Date:
February 08, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MOUNT TAM THERAPEUTICS INC (US)
International Classes:
A61K31/436; A61K31/453; A61K45/06
Domestic Patent References:
WO2013093493A12013-06-27
Foreign References:
US9382266B22016-07-05
US20080027089A12008-01-31
US20060205757A12006-09-14
Attorney, Agent or Firm:
HALLORAN, Patrick J. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for preventing and/or treating a neurodegenerative condition in a mammal, the method comprising administering to the mammal a compound of Formula I:

Formula I

wherein:

R is selected from H or -C(0)(CR3R4)b(CR5R6)d(CR7R8 R9);

R3 and R4 are each, independently, hydrogen, C1 to C6 alkyl, C2 to C8 alkenyl, C2 to C8 alkynyl, trihalomethyl, or -F;

R5 and R6 are each, independently, hydrogen, C1 to C6 alkyl, C2 to C8 alkenyl, C2 to C8 alkynyl, -(CR3R4)fOR10, -CFs, -F, or C02Rn;

R7 is hydrogen, C1 to C6 alkyl, C2 to C8 alkenyl, C2 to C8 alkynyl, -(CR3R4)fOR10, -CF3, -F, or CO2R11;

R8 and R9 are each, independently, hydrogen, C1 to C6 alkyl, C2 to C8 alkenyl, C2 to C8 alkynyl, -(CR3R4)fOR10, -CF3, -F, or CO2R11, or R8 and R9 can be taken together to form X or a cycloalkyl ring of 3-8 carbon atoms that is optionally mono-, di-, or tri-substituted with -(CR3R4)fOR10;

R10 is hydrogen, C1 to C6 alkyl, C2 to C8 alkenyl, C2 to C8 alkynyl, tri-(C1 to C6 alkyl)silyl, tri-(C1 to C6 alkyl)silylethyl, triphenylmethyl, benzyl, C2 to C8 alkoxymethyl, tri-(C1 to C6 alkyl)silylethoxymethyl, chloroethyl, or tetrahydropyranyl;

R11 is hydrogen, C1 to C6 alkyl, C2 to C8 alkenyl, C2 to C8 alkynyl, or a C7 to C10 phenylakyf

X is 5-(2,2-di-(Ci to C6 alkyl)[l,3]dioxanyl, 5-(2,2-di-(C3 to C8 cycloalkyl)[l,3]dioxanyl, 4-(2,2-di-(Ci to C6 alkyl)[l,3]dioxanyl, 4-(2,2-di-(C3 to C8 cycloalkyl)[l,3]dioxanyl, 4-(2,2-di-(C1 to C6 alkyl)[l,3]dioxalanyl, or 4-(2,2- di-(C3 to C8 cycloalkyl)[l,3]dioxalanyl;

b is a whole number from 0 to 6;

d is a whole number from 0 to 6; and, f is a whole number from 0 to 6; and/or,

a pharmaceutically acceptable salt, solvate, ester, or mixture thereof.

2. The method of claim 1 wherein the compound of Formula I is compound of Formula II:

Formula

and/or a pharmaceutically acceptable salt, solvate, ester, or mixture thereof.

3. The method of claim 1 or 2 wherein the neurodegenerative condition is selected from the group consisting of Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), motor neurone disease (MND), Lou Gehrig's Disease, Batten disease, choreic syndrome, dystonic syndrome, Friedrich's ataxia, Fluntington's disease, Lewy body disease, primary or secondary Parkinson's disease, prion disease, spinocerebellar ataxia, spinal muscular dystrophy, aging, alcoholism, cancer, injury, stroke, infectious disease, psychological disorders, and toxicity.

4. The method of any preceding claim wherein the neurodegenerative condition is selected from the group consisting of Alzheimer's disease, multiple sclerosis, Parkinson's disease, and Huntington's disease.

5. The method of any preceding claim wherein the compound of Formula I and/or Formula II is administered as the sole active pharmaceutical agent; or the compound of Formula I and/or Formula II is administered in combination with one or more agents selected from the group consisting of analgesics, anti- amyloidogenic, anti-cancer agents, anti-cholinergic agents, anti-convulsives, anti-depressants, anti-oxidants, anti-psychotics, anxiolytics, cell cycle inhibitors, dopamine releasing agents, dopamine replenishing agents, dopaminergic agonists, immunomodulatory, anti-inflammatory, immunostimulant, metabolic modulator, neuroprotectants, nootropic agents, vasoprotectants, and vasodilatory agents.

6. The method of any preceding claim wherein the administration is via a route selected from the group consisting of parenteral, oral, topical, buccal, sublingual, transdermal, a medical device, a stent, inhalation, injection, subcutaneous, intramuscular, or intravenous; wherein the administration comprises a single dose or multiple doses at the same or different dosages; and/or the members of a combination are administered physically and/or temporally simultaneously or separately.

7. The method of any preceding claim wherein the compound is provided as a bead, tablet, capsule, solution, or suspension.

8. Use of a compound of Formula I and/or Formula II in the preparation of a medicament for the prevention and/or treatment of a neurodegenerative condition.

9. The use of claim 8 wherein the neurodegenerative condition is selected from the group consisting of Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), motor neurone disease (M ND), Lou Gehrig's Disease, Batten disease, choreic syndrome, dystonic syndrome, Friedrich's ataxia, Fluntington's disease, Lewy body disease, primary or secondary Parkinson's disease, prion disease, spinocerebellar ataxia, spinal muscular dystrophy, aging, alcoholism, cancer, injury, stroke, infectious disease, psychological disorders, and toxicity.

10. The use of claim 8 or 9 wherein the compound of Formula I and/or Formula II is administered as the sole active pharmaceutical agent; or the compound of Formula I and/or Formula II is administered in combination with one or more agents selected from the group consisting of analgesics, anti-amyloidogenic, anti-cancer agents, anti-cholinergic agents, anti-convulsives, anti-depressants, anti-oxidants, anti-psychotics, anxiolytics, cell cycle inhibitors, dopamine releasing agents, dopamine replenishing agents, dopaminergic agonists, immunomodulatory, anti-inflammatory, immunostimulant, metabolic modulator, neuroprotectants, nootropic agents, vasoprotectants, and vasodilatory agents.

11. The use of any one of claims 8-10 wherein the administration is via a route selected from the group consisting of parenteral, oral, topical, buccal, sublingual, transdermal, a medical device, a stent, inhalation, injection, subcutaneous, intramuscular, or intravenous; wherein the administration comprises a single dose or multiple doses at the same or different dosages; and/or the members of a combination are administered physically and/or temporally simultaneously or separately.

12. The use of any one of claims 8-11 wherein the compound of Formula I is provided as a bead, tablet, capsule, solution, or suspension.

13. A kit for preventing and/or treating a neurodegenerative condition, the kit comprising at least one therapeutically effective dose of the compound of Formula I and/or Formula II, and instructions for preventing and/or treating a neurodegenerative condition using the same.

14. The kit of claim 13 wherein the instructions refer to administration is via a route selected from the group consisting of parenteral, oral, topical, buccal, sublingual, transdermal, a medical device, a stent, inhalation, injection, subcutaneous, intramuscular, or intravenous; wherein the administration comprises a single dose or multiple doses at the same or different dosages; and/or the members of a combination are administered physically and/or temporally simultaneously or separately.

15. The kit of claim 13 or 14 wherein the compound of Formula I is provided as a bead, tablet, capsule, solution, or suspension.

1 /12

Rapamycin numbering scheme

FIG.1

G 2A FI.

(S0l|onoi |B101%) S0L|OnOl |BJ0}B|BJ1UOO

4/12

(seipnoi |BJOI%) segonoj |BJ01B|BJJUOO

6/12

Vehicle Rapamycin Formula II

FIG.3B Z

ggW

v«t

■.■Si' A

1111

'IH»

§11»

»;

§¾§.¾

8/12

Vehicle Rapamycin Formula II

FIG.4B 9/12

10/12

(%) NS U! S|8A9-| HI

11/12

12/12

(%) ndO U! S|0A0| Hi

Description:
RAPAMYCIN ANALOG FOR PREVENTION AND/OR TREATMENT OF

NEURODEGENERATIVE CONDITIONS

RELATED APPLICATIONS

[0001] This application claims priority to U.S. Ser. No. 62/628,476 filed on February 9, 2018.

BACKGROUND OF THE DISCLOSURE

[0002] Rapamycin (sirolimus) is a polyketide that is used to coat coronary stents and prevent organ transplant rejection. The art also suggests that rapamycin and rapamycin analogs can be used to treat lymphangioleiomyomatosis, pulmonary inflammation (U.S. Patent 5,080,999), insulin dependent diabetes (U.S. Patent 5,362,718 citing Fifth Int. Conf. Inflamm. Res. Assoc. 121 (Abstract), (1990)), certain coronary diseases (Morris, (1992) Fleart Lung Transplant 11:197), leukemia and lymphoma (European Patent Application 0 525 960), and ocular inflammation (European Patent Application 0 532 862).

[0003] Rapamycin is produced by Streptomyces hygroscopicus NRRL 5491 (Sehgal et al., 1975; Vezina et al., 1975; U.S. 3,929,992; U.S. 3,993,749). For the purpose of this disclosure, rapamycin is described by the numbering convention of McAlpine et al. (1991) (see Fig. 1) in preference to the numbering conventions of Findlay et al. (1980) or Chemical Abstracts (11th Cumulative Index, 1982-1986 p60719CS). U.S. Patent 5,362,718 discloses acylated prodrugs of rapamycin.

[0004] Rapamycin and its commercially available analogs Temsirolimus and Everolimus inhibit activation of T cells and B cells by binding to mTOR which, among other things, reduces the production of interleukin-2. mTOR is the catalytic subunit of two structurally distinct complexes: mTORCl and mTORC2 (Wang et al. (2006) Journal of Biological Chemistry, 281: 24293-303). mTORCl and mTORC2 localize to different subcellular compartments, which affects their activation and function.

[0005] Scientific evidence suggests that mTORCl functions as a sensor of cellular nutritional and energy status and has a role in the regulation of protein synthesis (Hay et al. (2004) Genes & Development 18: 1926-45; Kim et al. (2002) Cell, 110: 163-75). The activity of mTORCl is regulated by rapamycin analogs, insulin, growth factors, phosphatidic acid, some amino acids and amino acid derivatives, mechanical stimuli, and oxidative stress.

[0006] Scientific evidence suggests that mTORC2 functions an important regulator of the actin cytoskeleton through its stimulation of F-actin stress fibers, paxillin, RhoA, Racl, Cdc42, and protein kinase Ca (Sarbassov et al. (2004) Current Biology 14:1296-302). mTORC2 also affecting metabolism and survival apparently through phosphorylation of Akt/PKB (Betz et al. (2013) PNAS 110: 12526-34). Akt phosphorylation by mTORC2 interacts with PDK1 and leads to full Akt activation (Sarbassov et al. (2005) Science 307: 1098-101; Stephens et al. (1998) Science 279: 710-4). In addition, mTORC2 is capable of activating IGF-IR and InsR (Yin et al. (2016) Cell Research 26: 46-65).

[0007] Rapamycin analogs (including Rapamycin) have significant therapeutic value (Huang et al, 2003). These polyketides are a potent inhibitor of the mammalian target of rapamycin (mTOR), a serine-threonine kinase downstream of the phosphatidylinositol 3-kinase (PI3K)/Akt (protein kinase B) signaling pathway that mediates cell survival and proliferation. This inhibitory activity is gained after rapamycin binds to the immunophilin FK506 binding protein 12 (FKBP12) (Dumont, F. J. and Q. X. Su, 1995). In T cells, rapamycin inhibits signaling from the IL-2 receptor and subsequent autoproliferation of the T cells resulting in immunosuppression. Rapamycin is marketed as an immunosuppressant for the treatment of organ transplant patients to prevent graft rejection (Huang et al, 2003). In addition to immunosuppression, rapamycin has found therapeutic application in cancer (Vignot et al, 2005).

[0008] Rapamycin and many rapamycin analogs have disadvantages including inducement of hyperlipidemia, cellular efflux mediated by P-glycoprotein ("P-gp"; LaPlante et al, 2002, Crowe et al, 1999) and other efflux mechanisms which pumps the compound out of cells and tends to decrease effectiveness of administered drug compound and presents challenges to the treatment of multidrug resistant cancer. Hepatic first pass loss of rapamycin is also high, which contributes further to its low oral bioavailability. The low oral bioavailability of rapamycin causes significant inter-individual variability resulting in inconsistent therapeutic outcome and difficulty in clinical management (Kuhn et al, 2001, Crowe et al, 1999).

[0009] A wide range of synthesized rapamycin analogues using the chemically available sites of the molecule are known in the art. Chemically available sites on the molecule for derivatization or replacement are known in the art to include, for example, C40 and C28 hydroxyl groups (e.g. U.S. 5,665,772; U.S. 5,362,718); C39 and C16 methoxy groups (e.g. WO 96/41807; U.S. 5,728,710); C32, C26 and C9 keto groups (e.g. U.S. 5,378,836; U.S. 5,138,051; U.S. 5,665,772); hydrogenation at C17, C19 and/or C21 (e.g. U.S. 5,391,730; U.S. 5,023,262); and/or the formation of oximes at C32, C40 and/or C28, (e.g., U.S. 5,563,145, U.S. 5,446,048). Analogues exhibiting resistance to metabolic attack (e.g. U.S. 5,912,253); bioavailability (e.g. U.S. 5,221,670; U.S. 5,955,457; WO 98/04279); and/or the production of prodrugs (e.g. U.S. 6,015,815; U.S. 5,432,183) have also been developed. Thus, it is understood in the art that the number of pharmaceutically useful and interesting analogs of rapamycin is very high and difficult to quantify.

[00010] While not intending to be bound be theory, we believe that rapamycin-like polyketide inhibitors of mTOR having a more balanced (e.g., less selective) ability to inhibit mTORCl and mTORC2 are preferred for the treatment of neurodegenerative disease and graft rejection because this permits a higher therapeutic index (i.e., more effective treatment of the disease or condition with lesser drug-induced adverse side effects). This disclosure provides a rapamycin analog that is a stereoisomer of the compound described in U.S. Pat. No. 9,382,266. U.S. Pat. No. 9,382,266 does not disclose or suggest the compounds of Formula I or II provided in the present disclosure, provide a composition in which the majority of the polyketide in the composition is the compound of Formula I or Formula II, or disclose or suggest the use of a compound of Formula I or Formula II for the prevention and/or treatment of neurodegenerative conditions.

SUMMARY OF THE DISCLOSURE

[00011] In some embodiments, this disclosure provides a polyketide of Formula I or Formula II that has an unexpected and beneficial pharmaceutical uses such as in the prevention and/or treatment of neurodegenerative conditions. In some embodiments, this disclosure provides compositions comprising the polyketide of Formula I or Formula II for use in preventing and/or treating neurodegenerative conditions. In some embodiments, this disclosure provides methods of preventing and/or treating neurodegenerative conditions in a mammal in need thereof comprising administration of the polyketide of Formula I and/or Formula II, and/or compositions and/or mixtures comprising the same. Other embodiments are also contemplated as described herein and/or as may be ascertained by those of ordinary skill in the art.

BRIEF DESCRIPTION OF THE DRAWINGS

[00012] Figure 1: The rapamycin numbering schema used in this document.

[00013] Figure 2. Effect of the compound of Formula II on contralateral touches in mice. Fig. 2A. Image of mice within testing device. Fig. 2B. Percent total touches (10 mg/kg). Fig. 2C. Percent total touches (3 mg/kg).

[00014] Figure 3. Effect of the compound of Formula II on integrated density in striatum. Fig. 3A. Image analysis. Fig. 3B. Percent of control side.

[00015] Figure 4. Effect of the compound of Formula II on TH + neuronal loss. Fig. 4A. Image analysis. Fig. 4B. TH + neuronal loss as percent of control side. Fig. 4C. TH staining. Fig. 4D. TH levels in SN (%). Fig. 4E. TH staining. Fig. 4F. TH levels in CPU (%). DETAILED DESCRIPTION

[00016]This disclosure provides the polyketides of Formula I and Formula II, which are C37-[(lR,2S,4R,5S)-5- hyhroxybicyclo[2.2.1]heptane] rapamycin and prodrug esters thereof (as described in U.S. Serial No. 62/457,676, the disclosure of which is incorporated herein into this disclosure in its entirety) for use in preventing and/or treating neurodegenerative conditions. Compounds of Formula I are prodrugs of the compounds of Formula II which, as described herein, have surprising and unexpectedly beneficial properties for the treatment of mammalian diseases. Formula I is shown below:

wherein:

R is selected from hydrogen, or -C(O)(CR 3 R 4 ) b (CR 5 R 6 ) d (CR 7 R 8 R 9 );

R 3 and R 4 are each, independently, hydrogen, C 1 to C 6 alkyl, C 2 to C 8 alkenyl, C 2 to C 8 alkynyl, trihalomethyl, or -F;

R 5 and R 6 are each, independently, hydrogen, C 1 to C 6 alkyl, C 2 to C 8 alkenyl, C 2 to C 8 alkynyl, - (CR 3 R 4 ) f OR 10 , -CFs, -F, or C0 2 R ;

R 7 is hydrogen, C 1 to C 6 alkyl, C 2 to C 8 alkenyl, C 2 to C 8 alkynyl, -(CR 3 R 4 ) f OR 10 , -CF 3 , -F, or CO 2 R 11 ;

R 8 and R 9 are each, independently, hydrogen, C 1 to C 6 alkyl, C 2 to C 8 alkenyl, C 2 to C 8 alkynyl, - (CR 3 R 4 ) f OR 10 , -CF 3 , -F, or CO 2 R 11 , or R8 and R9 can be taken together to form X or a cycloalkyl ring of 3-8 carbon atoms that is optionally mono-, di-, or tri-substituted with -(CR 3 R 4 ) f OR 10 ;

R 10 is hydrogen, C 1 to C 6 alkyl, C to C 8 alkenyl, C to C 8 alkynyl, tri-(C 1 to C 6 alkyl)silyl, tri-(C 1 to C 6 alkyl)silylethyl, triphenylmethyl, benzyl, C to C 8 alkoxymethyl, tri-(C 1 to C 6 alkyl)silylethoxymethyl, chloroethyl, or tetrahydropyranyl;

R 11 is hydrogen, C 1 to C 6 alkyl, C 2 to C 8 alkenyl, C 2 to C 8 alkynyl, or a C 7 to C 10 phenylakyf X is 5-(2,2-di-(C 1 to C 6 alkyl)[l,3]dioxanyl, 5-(2,2-di-(C 3 to C 8 cycloalkyl)[l,3]dioxanyl, 4-(2,2-di-(C 1 to C 6 alkyl)[l,3]dioxanyl, 4-(2,2-di-(C 3 to C 8 cycloalkyl)[l,3]dioxanyl, 4-(2,2-di-(C 1 to C 6 alkyl)[l,3]dioxalanyl, or 4-(2,2- di-( C 3 to C 8 cycloalkyl)[l,3]dioxalanyl;

b is a whole number from 0 to 6;

d is a whole number from 0 to 6; and,

f is a whole number from 0 to 6.

In a preferred embodiment, R contains at least one moiety selected from -(CR 3 R 4 ) f OR 10 , X or -(CR 3 R 4 )fOR 10 substituted C 3 to C 8 cycloalkyl. Pharmaceutically acceptable salts of such compounds are also provided.

[00017] The prodrugs of Formula I are convertible upon administration to a suitable mammal to the compound of Formula II. In some embodiments, the area under the curve formed by a plot of the concentration of the moiety of Formula I administered versus time is less than the area under the curve formed by a plot of the concentration of the compound (or compounds) of Formula II versus time. In some embodiments, the prodrug of formula II is at least 10-fold, and preferably at least 100-fold less pharmaceutically active than a compound of Formula II. In some embodiments, at least 10%, and preferably at least 50%, and more preferably at least 85% of the compound of Formula I is converted to the compound of Formula II during the time following administration to a mammal of the compound of Formula I that is equivalent to the biological half-life of the administered compound of Formula I. In some of the foregoing embodiments, the compound of Formula I is substantially pharmaceutically inert until conversion into the compound of Formula II. Flowever, in other embodiments, the compound of Formula I is significantly pharmaceutically active prior to conversion into the compound of Formula II.

[00018] In a preferred embodiment, the polyketide of Formula I is the polyketide of Formula II:

[00019]The polyketide disclosed herein, e.g., that of Formulas I or II, despite having structural relatedness to the polyketide disclosed in US Patent No. 9,382,266, rapamycin, and other analogs of rapamycin, shows a surprising and unexpectedly advantageous pharmacological profile as compared thereto. For instance, the polyketide of Formula II has unexpected advantages for the treatment of certain medical conditions to the polyketide disclosed in US Patent No. 9,382,266. Other advantages are indicated in Table 1.

Table 1

[00020]The polyketide prodrugs of Formula I, and in particular the polyketide of Formula II, also have unexpectedly beneficial pharmacokinetics. In particular, the polyketide of Formula II has a high oral bioavailability measured at around 0.47 (%F). This high oral bioavailability is substantially and significantly better than the polyketide disclosed in U.S. Patent No. 9,382,266 which is about one-half to about one-quarter lower than 0.47 (%F). In an aspect of the present invention, this comparative bioavailability can permit pharmaceutically effective administration of the composition of Formula II with lower toxicity (i.e., an increased therapeutic window). In another aspect of the present invention, this increased bioavailability improves the ability to orally administer the compound of Formula II, relative to the ability to orally administer the polyketide disclosed in U.S. Patent No. 9,382,266. The benefits of oral administration relative to intravenous and other routes of administration are well understood in the art.

[00021] The compounds described herein (e.g., a compound of Formula I, preferably a compound of

Formula II, and compositions comprising the same) can be used as a medicament for the prevention and treatment of various neurodegenerative conditions (e.g., a neurodegenerative disease) such as those resulting from the progressive loss of structure or function of neurons, including death thereof. Exemplary neurodegenerative conditions (e.g., a neurodegenerative disease) include, but are not limited to, Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS) (motor neurone disease (MND), Lou Gehrig's Disease), Batten disease, choreic syndrome, dystonic syndrome, Friedrich's ataxia, Fluntington's disease, Lewy body disease, primary or secondary Parkinson's disease, prion disease, spinal cerebellar ataxis, spinal muscular dystrophy, in mammals (including human). Other conditions that can lead to or result from neurodegeneration and may benefit from administration of the compounds described herein (e.g., a compound of Formula I, preferably a compound of Formula II, and compositions comprising the same) can include, for example, aging, alcoholism, cancer, injury, stroke, infectious disease (e.g., prion disease), psychological disorders, and/or toxicity (e.g., brain iron accumulation). Symptoms of neudegeneration, such as Parkinson's disease, can include one or more of bradykinesia, or slowness in voluntary movement, which produces difficulty initiating movement as well as difficulty completing movement once it is in progress, tremors in the hands, fingers, forearm, or foot, muscle rigidity, stiff muscles, muscle pain, an expressionless, mask-like face, poor balance, and/or falls. Other neurodegenerative conditions (e.g., a neurodegenerative disease) have symptoms well-known to those of skill in the art, and may be prevented, treated and/or ameliorated by administration the compounds described herein (e.g., a compound of Formula I, preferably a compound of Formula II, and compositions comprising the same). Administration of a therapeutically effective amount of these compounds may improve any one or more of such symptoms following administration of a therapeutically effective amount thereof to a mammal exhibiting the same.

[00022]The efficacy of the compounds of the invention (e.g., a compound of Formula I, preferably a compound of Formula II, and/or compositions comprising the same) may be tested in in vivo models for neurodegenerative conditions (e.g., a neurodegenerative disease) which are described herein and which are known to a person of skill in the art. Such models include, but are not limited to, for Alzheimer's disease— animals that express human familial Alzheimer's disease (FAD) p-amyloid precursor (APP), animals that overexpress human wild-type APP, animals that overexpress p-amyloid l-42(pA), animals that express FAD presenillin-1 (PS-1) (e. g. German and Eisch, 2004). For multiple sclerosis— the experimental autoimmune encephalomyelitis (EAE) model (see Bradl, 2003). For Parkinson's disease— the l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) model or the 6- hydroxydopamine (6-OHDA) model (see e.g. Emborg, 2004; Schober A. 2004). For Fluntington's disease there are several models including the R6 lines model generated by the introduction of exon 1 of the human Huntington's disease (HD) gene carrying highly expanded CAG repeats into the mouse germ line (Sathasivam et al, 1999) and others (see Hersch and Ferrante, 2004). Other models for testing the efficacy of the compounds described herein (e.g., a compound of Formula I, preferably a compound of Formula II, and compositions comprising the same) are also available to those of skill in the art, and may be suitable for determining the efficacy of these compounds.

[00023]The compound of Formula I or Formula II, or compositions comprising the same, can be administered via any suitable route or means including, but not limited to, parenterally, orally, topically (including buccal, sublingual, or transdermally), via a medical device such as a stent, by inhalation, or via injection (e.g., subcutaneously, intramuscularly, or intravenously). The treatment optionally consists of a single dose, but preferably in many embodiments is a multiplicity of administrations over time, at the same or different dosages. The skilled artisan will recognize that the optimal quantity and spacing of individual dosages of a compound of Formula I and/or Formula II will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the age and condition of the particular subject being treated, and that a physician will ultimately determine appropriate dosages to be used. This dosage may be repeated as often as appropriate. If side effects develop the amount and/or frequency of the dosage can be altered or reduced, in accordance with normal clinical practice.

[00024] In some embodiments, the compound of Formula I or Formula II is administered as the sole active pharmaceutical agent for preventing and/or treating neurodegenerative conditions. Thus, in some embodiments, a compound of Formula I or Formula II, or compositions comprising the same, is administered to a mammal such as a human being to prevent and/or treat a neurodegenerative condition as a pharmaceutical composition that optionally contains one or more pharmaceutical excipients, but no other active agent(s). In some embodiments, a compound of Formula I or Formula II, or a composition comprising the same, is administered to a mammal such as a human being to prevent and/or treat a neurological condition as a pharmaceutical composition comprising at least one other active agent, and optionally also contains one or more pharmaceutical excipients. In some embodiments, the compound of Formula I or a composition comprising the same is administered to a mammal such as a human being to prevent and/or treat a neurological condition as a pharmaceutical composition optionally comprising at least one other active agent, and optionally also containing one or more pharmaceutical excipients, with at least one other composition also comprising at least one other active agent and optionally also containing one or more pharmaceutical excipients (e.g., two compositions, one containing at least a compound of Formula I or Formula II, and the other composition comprising at least one other active agent). Multiple compositions, each comprising one or more active agents (one of such compositions comprising a compound of Formula I or Formula II), may also be administered to prevent and/or treat neurodegenerative disease. Such active agents and/or compositions, or some combination thereof, may be administered simultaneously or sequentially, and/or may be administered at the same or different sites on the mammal, and/or through the same or different routes of administration.

[00025] Active agents that may be administered to a mammal in order to prevent and/or treat a neurodegenerative condition along with a compound of Formula I or Formula II, or a composition comprising the same, include but are not limited to one or more analgesics, anti-amyloidogenic, anti-cancer agents, anti cholinergic agents, anti-convulsives, anti-depressants, anti-oxidants, anti-psychotics, anxiolytics, cell cycle inhibitors, dopamine releasing agents, dopamine replenishing agents, dopaminergic agonists, immunomodulator (e.g., anti-inflammatory, immunostimulant), metabolic modulator, neuroprotectants, nootropic agents, vasoprotectants (e.g., vasodilatory agents), and the like. Exemplary active agents that may be administered with along with the compounds described herein (e.g., a compound of Formula I, preferably the compound of Formula II) may include, for instance, any one or more of levodopa, carbidopa (SINEMET ® ), benserazide (MADOPAR ® , MADOPAR-FIBS), biperiden, catechol-O-methyltransferase (COMT) inhibitors (e.g., tolcapone, entacapone), dopamine agonists (e.g,. bromocriptine, pergolide, ropinirole, pramipexole, lisuride, cabergoline, apomorphine, sumanirole, rotigotine, talipexole and dihydroergocriptine), selegiline, trihexyphenidyl, a vaccine, and the like, and/or combinations and/or mixtures thereof, optionally along with any other active agents described herein or that may be otherwise available to those of skill in the art. A compound of Formula I or Formula II, or a composition comprising the same may also be administered in conjunction with any one or more of surgery, gene therapy, and the like, and/or any combinations thereof, optionally along with any of the active agents and/or procedures described herein or that may be otherwise available to those of skill in the art.

[00026] Compositions comprising a compound of Formula I or Formula II are optionally, but need not be, pure. A compound of Formula I or Formula II can be present in mixtures in which essentially all of the polyketide in the mixture is the polyketide of Formula I, and preferably Formula II, in which 99.9 % by weight of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II, in which 99.5% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II, in which at least 99% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II, in which 98% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II in which at least 95% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II, in which at least 90% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II, in which 80% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II, or in which the 70% of the polyketide in the mixture is the polyketide of Formula I or preferably Formula II.

[00027] Furthermore, in each of the foregoing aspects of the present invention (i.e., the compound of Formula I, and preferably Formula II), is optionally provided as a salt, a solvate, or an ester of the compound of Formula I, and preferably Formula II. Pharmaceutically acceptable salts of the polyketide of a compound of Formula I and/or Formula llinclude conventional salts formed from pharmaceutically acceptable inorganic or organic acids or bases as well as quaternary ammonium acid addition salts. More specific examples of suitable acid salts include hydrochloric, hydrobromic, sulfuric, phosphoric, nitric, perchloric, fumaric, acetic, propionic, succinic, glycolic, formic, lactic, maleic, tartaric, citric, palmitic, malonic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, fumaric, toluenesulfonic, methanesulfonic, naphthalene-2-sulfonic, benzenesulfonic hydroxynaphthoic, hydroiodic, malic, steroic, tannic and the like. Other acids such as oxalic, while not in themselves pharmaceutically acceptable, optionally can be useful in the preparation of salts useful as intermediates in obtaining the compounds of Formula I and/or Formula Hand their pharmaceutically acceptable salts. More specific examples of suitable basic salts include sodium, lithium, potassium, magnesium, aluminum, calcium, zinc, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N- methylglucamine and procaine salts. In an aspect of the present invention, pharmaceutically acceptable salts of the polyketide of Formula I or Formula II are combined together with one or more pharmaceutically acceptable excipients, diluents, or carriers.

[00028] Similarly, the polyketides of Formula I and/or Formula II, and pharmaceutically acceptable salts thereof, optionally can be solvates, including alcoholic solvates and hydrates.

[00029]The inventive polyketide (preferably a compound of Formula I or Formula II, most preferably Formula II) can be provided in a pure form for example in a crystalline or powdered form or diluted in at least one pharmaceutically acceptable buffer, carrier, or excipient. Pharmaceutically acceptable buffers, carriers and excipients in the context of the present invention preferably do not adversely interact with the polyketide of Formula I and/or Formula II, provide for stable formulations for suitable time periods, and are not unduly deleterious to most recipients thereof.

[00030] In some embodiments, solutions or suspensions of the compounds described herein (e.g., a compound of Formula I, preferably a compound of Formula II, and compositions comprising the same) also contain excipients such as, e.g., N,N-dimethylacetamide, dispersants e.g. polysorbate 80, surfactants, and solubilizers, e.g. polyethylene glycol, Phosal 50 PG (which consists of phosphatidylcholine, soya-fatty acids, ethanol, mono/diglycerides, propylene glycol and ascorbyl palmitate).

[00031]Tablets containing the inventive polyketide (preferably a compound of Formula I or Formula II, most preferably Formula II) optionally contain excipients such as microcrystalline cellulose, lactose (e.g. lactose monohydrate or lactose anyhydrous), sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, butylated hydroxytoluene (E321), crospovidone, hypromellose, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium, and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy- propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, and talc are optionally included.

[00032] Solid compositions of a similar type can also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the compounds of Formula I and/or Formula II can be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.

[00033] A tablet can be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g. povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets can optionally be coated or scored and can be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethylcellulose in varying proportions to provide desired release profile. [00034] For convenience, the formulations are optionally presented in unit dosage form and can be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient (compound of Formula I and/or Formula II) with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.

[00035] Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, impregnated dressings, sprays, aerosols or oils, transdermal devices, dusting powders, and the like. These compositions may be prepared via conventional methods containing the active agent. Thus, they may also comprise compatible conventional carriers and additives, such as preservatives, solvents to assist drug penetration, emollient in creams or ointments and ethanol or oeyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the composition. More usually they will form up to about 80% of the composition. As an illustration only, a cream or ointment is prepared by mixing sufficient quantities of hydrophilic material and water, containing from about 5-10% by weight of the compound, in sufficient quantities to produce a cream or ointment having the desired consistency.

[00036] Pharmaceutical compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. For example, the active agent may be delivered from the patch by iontophoresis.

[00037] For applications to external tissues, for example the mouth and skin, the compositions are preferably applied as a topical ointment or cream. When formulated in an ointment, the active agent may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active agent may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.

[00038] For parenteral administration, fluid unit dosage forms are prepared utilizing the active ingredient and a sterile vehicle, for example but without limitation water, alcohols, polyols, glycerine and vegetable oils, water being preferred. The active ingredient, depending on the vehicle and concentration used, can be either suspended or dissolved in the vehicle. In preparing solutions, the active ingredient can be dissolved in water for injection and filter sterilized before filling into a suitable vial or ampoule and sealing. [00039] Advantageously, agents such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle. To enhance the stability, the composition can be frozen after filling into the vial and the water removed under vacuum. The dry lyophilized powder is then sealed in the vial and an accompanying vial of water for injection may be supplied to reconstitute the liquid prior to use.

[00040] Parenteral suspensions are prepared in substantially the same manner as solutions, except that the active ingredient is suspended in the vehicle instead of being dissolved and sterilization cannot be accomplished by filtration. The active ingredient can be sterilized by exposure to ethylene oxide before suspending in the sterile vehicle. Advantageously, a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the active ingredient.

[00041]The compound of Formula I and/or Formula II may also be administered using medical devices known in the art. For example, in one embodiment, a pharmaceutical composition described herein can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Pat. No. 5,399,163; U.S. Pat. No. 5,383,851; U.S. Pat. No. 5,312,335; U.S. Pat. No. 5,064,413; U.S. Pat. No. 4,941,880; U.S. Pat. No. 4,790,824; or U.S. Pat. No. 4,596,556. Useful examples of well-known implants and modules include but are not limited to U.S. Pat. No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Pat. No. 4,486,194, which discloses a therapeutic device for administering medicaments through the skin; U.S. Pat. No. 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Pat. No. 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Pat. No. 4,439,196, which discloses an osmotic drug delivery system having multi-chamber compartments; and U.S. Pat. No. 4,475,196, which discloses an osmotic drug delivery system. In a specific embodiment, the polyketides (e.g., of Formula I or II) and compositions comprising the same may be administered using a drug-eluting stent, for example, such as one corresponding to those described in WO 01/87263 and related publications or those described by Perin (Perin, E C, 2005). Many other such implants, delivery systems, and modules are known to those skilled in the art.

[00042] As described herein, the polyketides and compositions described herein comprising a polyketide of Formula I, and/or preferably Formula II, can be administered to treat, prevent, or mitigate a neurodegenerative condition (e.g., disease or other neurodegenerative medical condition) in a mammal in need thereof. In a preferred embodiment, the mammal is a human. Any appropriate neurodegenerative condition of the mammal can be treated by administering a pharmaceutically-appropriate quantity of a compound of Formula I, and preferably of Formula II, or a composition comprising the same, to a mammal in need thereof. An ordinarily skill artisan can readily select the route of administration of a compound of Formula I, and preferably of Formula II, or a composition comprising the same, as well as the quantity following routine studies, guidelines and procedures. The dosage to be administered of a compound of Formula I, and preferably of Formula II, or a composition comprising the same, will vary according to the particular compound, the neurodegenerative condition involved, the subject, and the nature and severity of the neurodegenerative condition, the physical condition of the subject, and the selected route of administration. The appropriate dosage can be readily determined by a person skilled in the art. For example, without limitation, a dose of about 0.1 mg up to 100 mg daily, and optionally about 0.1 to 15 mg daily (or a higher dose given less frequently) can be administered. In some embodiments, a dose of about 1 or 3 mg/kg of Formula I i.v. or 1, 5 or 10 mg/kg of the compound of Formula I p.o., or a dose of from 1-20 mg/kg (e.g., about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, 19 or 20 mg/kg) can be administered one or more times.

[00043]The compositions may contain any suitable combination of a compound of Formula I or Formula II and other components. In some preferred embodiments, the compositions of the invention contain from 0.1 weight % to 70 weight % of the inventive polyketide (i.e., a compound Formula I and/or Formula II, preferably Formula II), preferably from 5-60 weight %, more preferably from 10 to 30 weight %, of the inventive polyketide, depending on the method of administration and other factors.

[00044]The polyketide of Formula I and/or Formula II described herein can be produced as a direct fermentation product, by feeding a starter acid of formula (III).

[00046] Suitable conditions for such a process are described in WO 2004/007709 (US 2005/0272132 Al) and WO 2006/016167 (US 2009/0253732 Al), the contents of which are incorporated by reference in their entirety. Specifically, a mutant strain of the rapamycin producing organism, Streptomyces hygroscopicus, that lacks the rapK gene and is called S. hygroscopicus ArapK (BIOT-4010; See, Example 1 of US Patent No. 9,382,266, the methods and materials of which are herein incorporated by reference) was generated. Other suitable production strains include S. hygroscopicus MG2-10 (pLL178), a derivative of S. hygroscopicus NRRL5491. The generation of S. hygroscopicus MG2-10 is described in example 2 of WO 2004/007709, and to generate a suitable production strain, this should be complemented with rapIJM NOQL, using an expression plasmid such pLL178 (as described in example 7 of WO 2006/016167). Fermentation of BIOT-4010, or a similar strain, such as S. hygroscopicus MG2-10 (pLL178) (WO 2004/007709, WO 2006/016167) in a suitable medium, such as (but not limited to) MD6, at a suitable temperature, such as 26° C, with addition of exo-(lR,2S,4R,5S)-5- hydroxybicyclo[2.2.1]heptane-2-carboxylate, typically at 24 hours is then sufficient for the production of the compound of Formula I and/or Formula II. Peak titers are observed between 3 and 8 days from inoculation. The acid form of compound of formula (II) is exo-(lR,2S,4R,5S)-5-hydroxybicyclo[2.2.1]heptane-2-carboxyli c acid.

[00047] Rapamycin producing strains include Streptomyces hygroscopicus, Actinoplates sp. N902-109 (See, Nishida et al (1995)) and Streptomyces sp. A 91-261402 (See, WO 94/18207). Other rapamycin producing strains are mentioned in WO 95/06649. The contents of WO 94/18207 and WO 95/06649 are incorporated in the present patent document by reference in their entirety.

[00048]The compound of Formula I and/or Formula II can be purified, for example, from other fermentation products, including but not limited to other polyketides, by any suitable conventional separation techniques, such as but not limited to, flash chromatography, preparative FIPLC, and/or crystallization.

[00049] Accordingly, in one aspect, the present invention provides a process for preparing a compound of Formula I and/or Formula II in substantially pure form comprises the steps of (i) feeding a starter acid of formula

to a rapamycin producing strain that has been genetically altered either to remove or inactivate the rapK gene, or in other embodiments, to remove or inactivate a gene encoding a chorismatase with function equivalent to that of the rapK gene product RapK (See, Andexer et al., 2011); and (ii) isolating and purifying the compound of Formula I and/or Formula II, preferably Formula II.

[00050] Compounds of Formula I can be prepared by acylation of the compound of Formula II using protected hydroxyl and polypro acids, alkoxy or polyalkoxy carboxylic acids that have been activated, followed by removal of the alcohol protecting groups if so desired. Several procedures for carboxylate activation are known in the art, but the preferred methods utilize carbodiimides, mixed anhydrides, or acid chlorides. For example, an appropriately substituted carboxylic acid can be activated as a mixed anhydride, with an acylating group such as 2,4,6-trichlorobenzoyl anhydride. Treatment of Formula II with the mixed anhydride under mildly basic condition provides the desired compounds. Alternatively, the acylation reaction can be accomplished with l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and dimethylaminopyridine.

[00051] Vulnerable hydroxyls of Formula II can be protected during the synthesis of Formula I through routine addition of a suitable protecting group such as tert-butyl dimethylsilyl protecting group, which at a later stage can be removed under mildly acidic conditions such as in a solution of acetic acid/water/TFIF. Deprotection is further described in U.S. Patent No. 5,118,678, which is hereby incorporated by reference. Alternative synthetic methods are provided by the analogy to the methods in U.S. Patent No. 5,120,842, which is hereby incorporated by reference.

[00052]The compounds of Formula I and Formula II can be purified by any suitable separation technology including, but not limited to, preparative-scale chromatography.

[00053]Thus, in some embodiments, this disclosure provides a compound of Formula I as described above, and/or a pharmaceutically acceptable salt, solvate, ester, or mixture thereof for use in preventing and/or treating a neurodegenerative condition. In some embodiments, this disclosure provides a composition comprising such a compound, pharmaceutically acceptable salt, solvate, ester, or mixture and, optionally, at least one pharmaceutically acceptable carrier. In some embodiments, this disclosure provides a prodrug of Formula II, wherein the prodrug is a polyketide of Formula I, as well as pharmaceutically acceptable salts, solvates, and hydrates of the compound of Formula I. In some embodiments, this disclosure provides a compound of Formula II as well as pharmaceutically acceptable salts, solvates, esters, or mixtures thereof, and/or compositions comprising the same (e.g., pharmaceutical compositions comprising a pharmaceutically acceptable carrier) for use in treating and/or preventing a neurodegenerative condition. In some embodiments, this disclosure provides for such use of a composition comprising about 70% or more, about 80% or more, about 90% or more (i.e., "substantially pure"), about 95% or more, or about 99% or more of a compound selected from the group consisting of the compound of Formula II, a pharmaceutically acceptable salt thereof, a solvate thereof, an ester thereof of the compound of formula I, and/or mixtures of the foregoing. In some embodiments, the composition for such use contains an essentially pure mixture, wherein an essentially pure mixture may contain trace amounts or pharmaceutically insignificant amounts of other polyketides, of a compound selected from the group consisting of the compound of Formula I and preferably Formula II, pharmaceutically acceptable salts, solvates, and esters of the compound of Formula I, and/or preferably Formula II, and mixtures of the foregoing. In some embodiments, this disclosure provides for the use a pharmaceutical composition comprising a pharmaceutically acceptable carrier, diluent or excipient and a polyketide of Formula I and/or preferably Formula II, wherein the pharmaceutically acceptable salt, solvate, and/or hydrate of the compound of Formula I and/or preferably Formula II comprises at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 98% of the polyketide component of the pharmaceutical composition in treating and/or preventing a neurodegenerative condition. In some embodiments of such uses, the compound of Formula II or Formula II is essentially the only polyketide in the pharmaceutical composition. In some embodiments of such uses, the prodrug of Formula I may be substituted for the polyketide of Formula II. In some embodiments, the solvate, if present, is a hydrate.

[00054] In some embodiments, this disclosure also provides methods for treating a mammal in need of prevention and/or treatment of a neurodegenerative condition, the method comprising administering to said mammal (e.g., a human being) an effective amount (e.g., a therapeutically effective amount) of the compound, pharmaceutically acceptable salt thereof, a solvate thereof, an ester thereof of the compound of Formula I or Formula II, preferably Formula II, and/or a composition and/or mixture comprising the same. In some embodiments, the method of treating a mammal in need thereof comprises administering to said mammal an effective amount of a compound of Formula II, pharmaceutically acceptable salt thereof, solvate thereof, ester thereof, or mixture thereof and/or comprising the compound of Formula II; and/or a composition comprising Formula II (e.g., a therapeutically effective amount) thereto. In some embodiments, the mammal has a neurodegnerative condition selected from the group consisting of but are not limited to, for Alzheimer's disease, multiple sclerosis, Parkinson's disease, and/or Fluntington's disease.

[00055] In some embodiments, the method comprises administration of a compound of Formula I or Formula II to a mammal at about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 mg/kg (e.g., about 10 mg/kg) to provide a therapeutically effective mean concentration of in the whole blood of the mammal for a sufficient period of time (e.g., up to six hours) following administration. In some embodiments, the method comprises administering the compound of Formula I or Formula II to the mammal multiple times in a single day and/or over several days, weeks, months, and/or years. In some embodiments, such administration results in a reduction in the symptoms of the neurodegenerative condition to, e.g., about any of 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of symptoms before treatment was started, or between any two points in time and/or doses. In some embodiments, a significant reduction in symptoms is observed and exhibits an adjusted P value of 0.0001 as determined by Dunnett's multiple comparison's test. [00056] Any mode of administration may be utilized. In some embodiments, the compound, composition and/or mixture is administered by application to an implantable medical device (e.g., a stent).

[00057] In some embodiments, then, this disclosure provides methods for preventing and/or treating a neurodegenerative condition in a mammal by administering a compound of Formula I and/or Formula II, a pharmaceutically acceptable salt, solvate, ester, or mixture thereof in a therapeutically effective amount, and/or a composition comprising the same. In some embodiments, the neurodegenerative condition includes but is not limited to Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS) (motor neurone disease (MND), Lou Gehrig's Disease), Batten disease, choreic syndrome, dystonic syndrome, Friedrich's ataxia, Fluntington's disease, Lewy body disease, primary or secondary Parkinson's disease, prion disease, spinocerebellar ataxia, spinal muscular dystrophy, aging, alcoholism, cancer, injury, stroke, infectious disease, psychological disorders, and toxicity. In some embodiments, the neurodegenerative condition is Alzheimer's disease, multiple sclerosis, Parkinson's disease, or Huntington's disease. In some embodiments, the compound of and/or Formula II, or compositions comprising the same, is administered as the sole active pharmaceutical agent. In some embodiments, the compound of Formula I and/or Formula II, or a composition comprising the same, is administered in combination with one or more agents selected from the group consisting of analgesics, anti-amyloidogenic, anti-cancer agents, anti-cholinergic agents, anti-convulsives, anti-depressants, anti oxidants, anti-psychotics, anxiolytics, cell cycle inhibitors, dopamine releasing agents, dopamine replenishing agents, dopaminergic agonists, immunomodulatory, anti-inflammatory, immunostimulant, metabolic modulator, neuroprotectants, nootropic agents, vasoprotectants, and vasodilatory agents. In some embodiments, the administration is via a route selected from the group consisting of parenteral, oral, topical, buccal, sublingual, transdermal, a medical device, a stent, inhalation, injection, subcutaneous, intramuscular, or intravenous; wherein the administration comprises a single dose or multiple doses at the same or different dosages; and/or the members of a combination are administered physically and/or temporally simultaneously or separately. In some embodiments, the compound of Formula I and/or Formula II is provided as a bead, tablet, capsule, solution, or suspension. Also provided are uses of a compound of and/or Formula II in the preparation of a medicament for the prevention and/or treatment of a neurodegenerative condition (e.g., as described above). In some embodiments, kits for preventing and/or treating a neurodegenerative condition, the kit comprising at least one therapeutically effective dose of the compound of Formula I and/or Formula II, and instructions for preventing and/or treating a neurodegenerative condition using the same are also provided. In some embodiments, the instructions refer to administration is via a route selected from the group consisting of parenteral, oral, topical, buccal, sublingual, transdermal, a medical device, a stent, inhalation, injection, subcutaneous, intramuscular, or intravenous; wherein the administration comprises a single dose or multiple doses at the same or different dosages; and/or the members of a combination are administered physically and/or temporally simultaneously or separately. In some embodiments, the kit provides the compound of Formula I and/or Formula II as a bead, tablet, capsule, solution, or suspension. Other embodiments are also contemplated as would be understood by those of skill in the art.

[00058] When the terms treat, prevent, and / or ameliorate or derivatives thereof are used herein in connection with a given treatment for a given condition, it is meant to convey that the treated patient either does not develop a clinically observable level of the condition at all, or develops it more slowly and/or to a lesser degree than he/she would have absent the treatment. These terms are not limited solely to a situation in which the patient experiences no symptom of the condition whatsoever. For example, a treatment will be said to have prevented the condition if it is given during exposure of a patient to a stimulus that would have been expected to produce a given manifestation of the condition, and results in the patient's experiencing fewer and/or milder symptoms of the condition than otherwise expected. For instance, a treatment can "prevent" symptoms where the mammal expresses less symptoms than would have been observed in the absence of treatment; it does not imply that the mammal must not exhibit any symptoms. Similarly, reduce, reducing, and reduction as used herein in connection with prevention, treatment and / or amelioration of a given condition by a particular treatment typically refers to a subject developing an infection more slowly or to a lesser degree as compared to a control or basal level (e.g., of symptoms) in the absence of a treatment.

[00059]The terms "about", "approximately", and the like, when preceding a list of numerical values or range, refer to each individual value in the list or range independently as if each individual value in the list or range was immediately preceded by that term. The terms mean that the values to which the same refer are exactly, close to, or similar thereto. As used herein, a subject or a host or a mammal is meant to be an individual. The subject can include mammals such as domesticated animals, such as cats and dogs, livestock (e.g., cattle, horses, pigs, sheep, and goats), laboratory animals (e.g., mice, rabbits, rats, guinea pigs) and birds. A mammal may also be a primate or a human. Optional or optionally means that the subsequently described event or circumstance can or cannot occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, the phrase optionally the composition can comprise a combination means that the composition may comprise a combination of different compounds or molecules or may not include a combination such that the description includes both the combination and the absence of the combination (i.e., individual members of the combination). The term "combined" or "in combination" or "in conjunction" may refer to a physical combination of agents that are administered together or the use of two or more agents in a regimen (e.g., administered separately, physically and / or in time) for treating, preventing and / or ameliorating a particular disease. Ranges may be expressed herein as from about one particular value, and/or to about another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent about or approximately, it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. Ranges (e.g., 90-100%) are meant to include the range perse as well as each independent value within the range as if each value was individually listed. Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps. All references referred to in this application, including patent and patent applications, are incorporated herein by reference info this disclosure in their entirety.

[00060] Certain embodiments are further described in the following examples. These embodiments are provided as examples only and are not intended to limit the scope of the claims in any way.

EXAMPLES

Example 1

[00061]This example illustrates one method for determining the pharmacokinetics and bioavailability of the compound of the Formula I and/or Formula II.

[00062]A person of skill in the art will be able to determine the pharmacokinetics and bioavailability of the compound of Formula I and/or Formula II using in vivo and in vitro methods known to a person of skill in the art, including but not limited to those described below and in Gallant-Flaidner et al, 2000 and Trepanier et al, 1998 and references therein. The bioavailability of a compound is determined by a number of factors, (e.g. water solubility, cell membrane permeability, the extent of protein binding and metabolism and stability) each of which may be determined by in vitro tests as described in the examples herein, it will be appreciated by a person of skill in the art that an improvement in one or more of these factors will lead to an improvement in the bioavailability of a compound. Alternatively, the bioavailability of the compound of Formula I and/or Formula llmay be measured using in vivo methods as described in more detail below, or in the examples herein.

[00063] In order to measure bioavailability in vivo, a compound may be administered to a test animal (e.g. mouse or rat) both intraperitoneally (i.p.) or intravenously (i.v.) and orally (p.o.) and blood samples are taken at regular intervals to examine how the plasma concentration of the drug varies over time. The time course of plasma concentration over time can be used to calculate the absolute bioavailability of the compound as a percentage using standard models. An example of a typical protocol is described below.

[00064] For example, mice or rats are dosed with 1 or 3 mg/kg of the compound of the invention (e.g,. a compound for Formula I or Formula II, i.v. or 1, 5 or 10 mg/kg of the compound of the invention p.o.). Blood samples are taken at 5 min, 15 min, 1 h, 4 h and 24 h intervals, and the concentration of the compound of Formula I and/or Formula II in the sample is determined via LCMS-MS. The time-course of plasma or whole blood concentrations can then be used to derive key parameters such as the area under the plasma or blood concentration-time curve (AUC— which is directly proportional to the total amount of unchanged drug that reaches the systemic circulation), the maximum (peak) plasma or blood drug concentration, the time at which maximum plasma or blood drug concentration occurs (peak time), additional factors which are used in the accurate determination of bioavailability include: the compound's terminal half-life, total body clearance, steady-state volume of distribution and F %. These parameters are then analyzed by non-compartmental or compartmental methods to give a calculated percentage bioavailability, for an example of this type of method see Gallant-Haidner et al, 2000 and Trepanier et al, 1998, and references therein. Bioavailability may be determined as described herein in order to carry out studies of the effects of the compounds of Formula I, preferably a compound of Formula II, in neurodegenerative disease models and/or humans.

[00065] Shown below are whole blood concentrations following administration of the compound of Formula II to mice. The compound of Formula II was administered to mice at 2 mg/kg or 10 mg/kg for three days and the concentration of the compound in whole blood determined. These determinations were made using protein precipitation, liquid chromatography (LC), and mass spectrometry (MS/MS). Ten mI aliquots of whole blood and matrix calibration standards were distributed in a 96-well plate; 10 mI aliquots of blank matrix for matrix blanks and control blanks were included as controls. Ten mI of water was added to each sample followed by vortexing. One hundred sixty ml of internal standard was added to each sample except the matrix blanks; 160 mI 70:30 water:acetonitrile (ACN) was added to matrix blanks. This was followed by a five-minute vortex at > 3500 rpm. One hundred fifty mI of the resultant supernatant was then transferred to a new 96-well plate and the samples blown to dryness at 35°C. The resultant product was then reconstituted with 90 mI ACN. LC was carried out using the equipment, conditions and calibration standards are shown in Tables 2 and 3. Table 2

Table 3

N/A = Not Applicable // NR = Not Reported

The results of these analyses are summarized in Tables 4 and 5.

Table 4

Table 5

AQL: Original value above quantitation limit (1000 ng/mL). Sample was diluted prior to re-analysis, but due to a sequence error the samples will be re-injected.

[00066] As shown above, Formula II exhibits sufficient concentrations in whole blood over time following administration at 2 mg/kg or 10 mg/kg via the intraperitoneal route (IP) once daily (QD) for three days. To carry out these assays using brain homogenates, 20 uL of samples and matrix calibration standards into wells of a 96- well plate, and 10 uL of blank matrix allocated for matrix blanks and control blanks. 160 uL internal standard was then added to all samples except matrix blanks, and 160 uL ACN added to matrix blanks. Samples were then vortexed for 5 minutes and centrifuge for 5 minutes at >3500 rpm. 150 uL of the supernatant was then transferred to a new 96-well plate, and the plates blown down to dryness at 35°C, reconstituted with 90uL of 70:30 watenacetonitrile (ACN) solution, covered and vortexed prior to analysis. LC and MS conditions were the same as described above for the whole blood analysis. A summary of the Calibration of Standard Back-Calculated Concentration of Formula I in brain homogenate is shown in Table 6.

Table 6

NA=not applicable; NR=not reported

[00067] Shown below are brain concentrations following administration of the compound of Formula I to mice. The compound of Formula II was administered to mice at 2 mg/kg or 10 mg/kg for three days and the concentration of the compound in brain determined. The results are summarized in Table 7:

Table 7

[00068] As shown above, Formula II exhibits sufficient concentrations in brain over time following administration at 2 mg/kg or 10 mg/kg via the intraperitoneal route (IP) once daily (QD) for three days.

Example 2

[00069]The efficacy of the compound of Formula I and/or Formula II may be tested in in vivo models for neurodegenerative conditions (e.g., a neurodegenerative disease) which are described herein and which are known to a person of skill in the art. Such models include, but are not limited to, for Alzheimer's disease— animals that express human familial Alzheimer's disease (FAD) p-amyloid precursor (APP), animals that overexpress human wild-type APP, animals that overexpress p-amyloid l-42(pA), animals that express FAD presenillin-1 (PS- 1) (e. g. German and Eisch, 2004). For multiple sclerosis— the experimental autoimmune encephalomyelitis (EAE) model (see Bradl, 2003). For Parkinson's disease— the l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) model or the 6-hydroxydopamine (6-OHDA) model (see e.g. Emborg, 2004; Schober A. 2004). For Huntington's disease there are several models including the R6 lines model generated by the introduction of exon 1 of the human Huntington's disease (HD) gene carrying highly expanded CAG repeats into the mouse germ line (Sathasivam et al, 1999) and others (see Hersch and Ferrante, 2004).

[00070] An analysis of the neuroprotective effects of the compound of Formula II on motor performance was performed using the 6-OHDA cylinder test (Fig. 2A) which evaluates spontaneous motor changes associated with dopamine depletion in the striatum of 6-OHDA injected mice. Mice were pre-treated with the compound of Formula II (in 2% EtOH, 40% PEG 400 in saline (PBS IX)) at a dose of 10 mg/kg, rapamycin (10 mg/kg) (in 2% EtOH, 40% PEG 400 in saline (PBS IX)), or the vehicle control (2% EtOH, 40% PEG 400 in saline (PBS IX)) for three days prior to stereotactic injection of 6-OHDA (80 μg) into one hemisphere of the brain. 6-OHDA-treated mice were placed in a glass container and the number times each mouse touched the glass wall with each forepaw following administration of vehicle only (n = 7), rapamycin (n = 10), or the compound of Formula II (n = 10) measured. Control mice were administered the vehicle control only. "Baseline" mice were treated with 6-OHDA, vehicle, rapamycin or the compound of Formula II after the baseline measurement was taken. The data is presented in Fig. 2B as mean and standard error of the mean (SEM). Statistical analyses as compared to baseline mice were performed using the one-way Anova test (*: p < 0.05; n.s.: not significant). Mice to which compound of Formula II was administered following 6-OHDA treatment exhibited a non-significant difference from baseline (mice that were not treated by 6-OHDA), while both the vehicle control and rapamycin exhibited significant differences from baseline (a decrease in contralateral touches). As shown therein, the compound of Formula II clearly outperformed rapamycin and the vehicle control by bringing the number of touches closer to that of baseline mice. The data presented in Fig. 2C was generated in the same manner as was performed to generate the data presented in Fig. 2B (except that Formula II or rapamycin was administered at 3 mg/kg and pre treatment prior to the first 6-OHDA administration was for two days), and supports the data presented in Fig. 2B (vehicle (n=6); rapamycin (n=7); Formula II (n=8)). Data is presented as mean and SEM. Statistical analyses were performed using One-way Anova test (*: p < 0.05; ***: p < 0.001. n.s.: not significant.)

[00071] Immunohistochemistry analysis using anti-tyrosine hydroxylase (TH) antibody was performed in mouse midbrain sections to quantify 6-OHDA-induced striatal denervation in mice injected with 6-OHDA and non- injected (control) sides of the brain. The integrated density of pixel intensity of TH stain covering the entire striatum and expressed as a percentage of TH loss relative to the control slide, thereby determining the effect of vehicle control (treatment negative control (n=7)), rapamycin (n=10) and the compound of Formula II (n=10) on 6-OFIA-induced denervation. Figure 3A provides a representative visual evidence that rapamycin and the compound of Formula II decreased 6-OHDA-induced denervation as compared to vehicle control. Figure 3A also provides representative visual evidence that the compound of Formula II also decreased 6-OHDA-induced denervation as compared to rapamycin. For instance, in the Vehicle panels, the 6-OHDA / vehicle-treated striatum side shows essentially no to very little anti-TH antibody staining. In contrast, both the 6-OHDA / rapamycin-treated and the 6-OHDA/ Formula ll-treated sides of the brain show increased staining with the anti- TFI antibody, as indicated by the dark shading in those figures. Figure 3B presents the mean and standard error of the mean (SEM) of the integrated density of anti-TH antibody staining in the striatum, as well as statistical analysis of the results for the groups of mice. As shown therein, the one-way ANOVA test showed a significant difference between vehicle control and rapamaycin (p<0.05) as well as between vehicle control and the compound of Formula II (p<0.001). Accordingly, the compound of Formula II was shown to protect mice from 6-OHDA-induced striatal denervation as compared to vehicle control as well as rapamycin. [00072] Immunohistochemistry analysis using anti-tyrosine hydroxylase (TH) antibody was also performed to quantify 6-OHDA-induced neuronal loss in mice injected with 6-OHDA and non-injected (control) sides of the brain. The total content of TH-positive somas was measured in midbrain sections covering the entire SN in the 6-OHDA injected and non-injected sides for each group of mice, and the effect of vehicle control (treatment negative control (n=7)), rapamycin (n=10) and the compound of Formula II (n=10) thereupon. Figure 4A provides a representative visual evidence that rapamycin and the compound of Formula II decreased 6-OHDA-induced neuronal loss as compared to vehicle control as well as rapamycin. Figure 4B presents the mean and standard error of the mean (SEM) as well as statistical analysis of the results. As shown therein, the one-way ANOVA test showed a significant difference between vehicle control and rapamaycin (p<0.001) as well as between vehicle control and the compound of Formula II (p<0.01). This staining data was confirmed by staining for TH protein expression in the 6-OHDA-treated striatum (Figures 4C, 4D, 4E and 4F (3 mg/kg dose of Formula I or rapamycin, two-day pre-treatment)). As shown in Figure 4D, for instance, TH protein expression in 6-OHDA- / Vehicle- treated striatum ("Injected" with 6-OHDA) showed a statistically significant difference from control striatum ("Non Injected" with 6-OHDA), but a non-statistically significant difference from 6-OHDA / rapamycin-treated ("Injected" with 6-OHDA) or 6-OHDA / Formula l-treated ("Injected" with 6-OHDA) striatum. Accordingly, the compound of Formula II was shown to protect mice from 6-OHDA-induced neuronal loss.

[00073] In sum, this data shows that the compound of Formula II provides neuroprotection from 6-OHDA- induced neurotoxicity. As the 6-OHDA model is generally accepted as representative of Parkinson's disease, those of skill in the art would understand the compound of Formula II to have anti-Parkinson's disease effects, i.e., to be potentially useful in treating and/or preventing Parkinson's Disease in mammals such as human beings.

[00074] While certain embodiments have been described in terms of the preferred embodiments, it is understood that variations and modifications will occur to those skilled in the art. Therefore, it is intended that the appended claims cover all such equivalent variations that come within the scope of the following claims.