CYCLODEXTRIN FOR THE TREATMENT OF LYSOSOMAL STORAGE DISEASES

The invention provides for methods of treating lysosomal storage disorders and/or reduction of non-cholesterol lipids, using cyclodextrin compounds, including in combination with other therapeutics, including vitamin E.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application claims priority of U.S. Provisional Application No. 61/679,668, filed on Aug. 3, 2012, the entirety of which is incorporated by reference herein.

FEDERALLY SPONSORED RESEARCH

This work was supported by the Federal Government. The Government has certain rights in this invention.

TECHNICAL FIELD

The invention provides for methods of treating lysosomal storage disorders and/or reduction of non-cholesterol lipids, using cyclodextrin compounds and cyclodextrin compounds in combination with other agents.

BACKGROUND OF THE INVENTION

Cyclodextrins (CD) are sugar molecules in a ring form. The alpha-CD (6 sugars), beta-CD (7 sugars) and gamma-CD (8 sugars) are commonly used cyclodextrins. The hydroxypropyl-beta cyclodextrin (HPβCD) has been approved for the pharmaceutical use as the drug excipient. Recent reports showed that beta-cyclodextrin including HPβCD and beta-methyl-cyclodextrin (MβCD) reduced cholesterol accumulation and neuronal cell loss in the mouse model of Niemann Pick Type C (NPC) disease. The life span of these NPC KO mice also increased 80 to 100% after the CD treatment. The similar positive results were obtained in the feline model of NPC disease. It was also reported that beta-CD increased exocytosis in primary NPC fibroblasts.

It has been recently found that delta-tocopherol increased the cholesterol efflux from NPC cells and reduced cholesterol accumulation. Enhancement of lysosomal exocytosis has been indicated as a therapeutic strategy for development of new treatment for all lysosomal storage diseases that are composed of 50 different diseases caused by the genetic mutations of genes for lysosomal proteins. The phenotypic changes in these diseases are accumulation of lipids, glycoprotein and/or other macromolecules in lysosomes and enlarged lysosome size in patient cells that may lead to cell malfunction and cell death in affected tissues.

SUMMARY OF THE INVENTION

In one aspect, the invention provides a method of treating a lysosomal storage disorder in a subject, comprising determining that the subject is in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and macromolecule accumulation, and administering to the subject in need thereof, an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another aspect, the invention provides a method of treating a lysosomal storage disorder in a subject, wherein the subject has been previously identified as being in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and macromolecule accumulation, comprising administering to said subject in need thereof an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another aspect, the invention provides a method of treating a lysosomal storage disorder in a subject, comprising the step of administering to the subject an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and an additional therapeutic agent.

In certain aspects, the invention provides a method of reducing non-cholesterol lipids or reduction of non-cholesterol dominant lipid and macromolecule accumulation in a subject, the method comprising administering to the subject a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof; and detecting the amount of lipid reduction.

In another aspect, the invention provides a pharmaceutical composition comprising a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and vitamin E, together with a pharmaceutically-acceptable carrier or excipient.

In another aspect, the invention provides a method of treating a subject suffering from a lysosomal storage disorder, comprising the use of the pharmaceutical composition as described above, in combination with another agent.

In one embodiment of any of the above aspects, the step of administering the cyclodextrin compound comprises administering the cyclodextrin compound to a subject such as a human in a dosage of between about 0.01 mg/Kg/day and 100 mg/Kg/day.

In another embodiment of any of the above aspects, the cyclodextrin compound is administered to a subject such as a human in an amount from about 0.5 mg/Kg to 8 mg/Kg, either in a single dose or per day.

In one embodiment of any of the above aspects, the cyclodextrin compound is administered to a subject such as a human in an amount of about 3 mg/Kg either in a single dose or per day. In a further embodiment of any one of the above aspects, the cyclodextrin compound is administered to a subject such as a human in an amount of about 1.0 mg/Kg, 1.25 mg/Kg, 1.5 mg/Kg, 1.75 mg/Kg, 2.0 mg/Kg, 2.25 mg/Kg, 2.5 mg/Kg, 2.75 mg/Kg, 3.25 mg/Kg, 3.5 mg/Kg, 3.75 mg/Kg, 4.0 mg/Kg, 4.25 mg/Kg, or 4.5 mg/Kg, either in a single dose or per day.

In another further embodiment of any of the above aspects, the cyclodextrin compound is administered to a subject such as a human in an amount from about 0.1 mg/Kg to 0.3 mg/Kg, 0.1 mg/Kg to 0.4 mg/Kg, 0.1 mg/Kg to 0.5 mg/Kg, 0.1 mg/Kg to 0.6 mg/Kg, or 0.1 mg/Kg to 0.7 mg/Kg, either in a single dose or per day.

In another further embodiment of any of the above aspects, the cyclodextrin compound is administered in a single dose.

In one embodiment of any of the above aspects, the additional therapeutic agent (distinct from the cyclodextrin compound) is administered to a subject such as a human in an amount from about 0.05 to 1 mg/kg, either in a single dose or per day. In another embodiment of any of the above aspects, the additional therapeutic agent (distinct from the cyclodextrin compound) is administered to a subject such as a human in an amount from about 0.1 mg/Kg to 0.3 mg/Kg, 0.1 mg/Kg to 0.4 mg/Kg, 0.1 mg/Kg to 0.5 mg/Kg, 0.1 mg/Kg to 0.6 mg/Kg, or 0.1 mg/Kg to 0.7 mg/Kg, either in a single dose or per day.

In one embodiment of any of the above aspects, the additional therapeutic agent is administered in a single dose.

In another aspect, the invention features a method of treating a lysosomal storage disorder in a subject, comprising determining that the subject is in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and macromolecule accumulation; administering to the subject in need thereof, an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof in an amount from about 0.05 mg/Kg to 1 mg/Kg either in a single dose or per day; and administering to the subject an additional therapeutic agent (such as vitamin E) distinct from the cyclodextrin compound in an amount from about 0.05 mg/Kg to 1 mg/Kg either in a single dose or per day.

In another aspect, the invention features a method of treating a lysosomal storage disorder in a subject, comprising determining that the subject is in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and macromolecule accumulation; administering to the subject in need thereof, an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof; and administering to the subject an additional therapeutic agent (such as vitamin E) distinct from the cyclodextrin compound.

In one embodiment, the cyclodextrin compound is administered to a subject such as a human in an amount from about 0.1 mg/Kg to 0.3 mg/Kg, 0.1 mg/Kg to 0.4 mg/Kg, 0.1 mg/Kg to 0.5 mg/Kg, 0.1 mg/Kg to 0.6 mg/Kg or 0.1 mg/Kg to 0.7 mg/Kg, either in a single dose or per day.

In another embodiment, the additional therapeutic agent (distinct from the cyclodextrin compound) is administered to a subject such as a human in an amount from about 0.1 mg/Kg to 0.3 mg/Kg, 0.1 mg/Kg to 0.4 mg/Kg, 0.1 mg/Kg to 0.5 mg/Kg, 0.1 mg/Kg to 0.6 mg/Kg or 0.1 mg/Kg to 0.7 mg/Kg, either in a single dose or per day.

In still another further embodiment, the cyclodextrin compound is administered to a subject such as a human in an amount of about 50 uM in combination with 10 uM of an additional agent, either in a single dose or per day.

In a further preferred embodiment, the cyclodextrin compound is administered to a subject such as a human in an amount of about 50 uM in combination with 10 uM of delta-tocopherol, either in a single dose or per day.

In other aspects, the invention provides a kit comprising an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in unit dosage form, together with instructions for administering the compound to a subject suffering from a lysosomal storage disorder.

Various advantages of the invention include the following: Treatment of all lysosomal storage diseases with cyclodextrins, including hydroxypropyl-beta-cyclodextrin, but in certain aspects with the exception of Niemann Pick Type C disease; treatment of all lysosomal storage diseases with cyclodextrins, including hydroxypropyl-beta-cyclodextrin in combination of vitamin-E, for synergistic or additive therapeutic effect, for reduction of dosage of cyclodextrin needed that makes the cyclodextrin treatment more practically feasible, and for less side effects by reducing dosages of both drugs; treatment of all lysosomal storage diseases with cyclodextrins (such as beta and gama forms) in combination with modified cyclodextrins for better efficacy and less side effects; treatment of all lysosomal storage diseases with cyclodextrins and modified vitamin-E analogs for the better efficacy and less side effects.

Further, the invention provides the administration of the compounds of the invention for the treatment of all 40-50 lysosomal storage diseases based on the mechanism of action of cyclodextrin (increases lysosomal exocytosis).

Other aspects of the invention are disclosed infra.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1 is a set of graphs showing the reduction of total cholesterol (including cholesterol ester and free cholesterol) in Wolman fibroblasts treated with δ-tocopherol, α-tocopherol, methyl-β-cyclodextrin, and combinations of δ-tocopherol and methyl-β-cyclodextrin, and α-tocopherol and methyl-β-cyclodextrin (MBCD).

FIG. 2 is a set of photographs showing Nile Red staining of Wolman fibroblasts treated with δ-tocopherol (D-T), α-tocopherol (a-T), methyl-β-cyclodextrin, and combinations of δ-tocopherol (D-T) and methyl-β-cyclodextrin (MBCD), and α-tocopherol and methyl-β-cyclodextrin.

FIG. 3 is a set of graphs showing exocytosis levels in Wolman fibroblasts as measured by HEXB secretion.

FIG. 4 is a set of graphs showing lysosomal calcium efflux in wild-type fibroblasts and lysosomal storage disease fibroblasts in the presence and absence of δ-tocopherol, α-tocopherol, methyl-β-cyclodextrin, and combinations of δ-tocopherol and methyl-β-cyclodextrin, and α-tocopherol and methyl-β-cyclodextrin.

FIGS. 5A-5D are sets of photographs showing the effect of various forms of Cyclodextrins (a-CD, r-CD), δ-Tocopherol (D-T), and combinations in seven disease and wild-type fibroblast cell lines as measured using the Lysotracker assay. Highly branched cyclodextrin (HBCD), also known as Kleptose.

FIG. 6 is a set of photographs showing that cyclodextrin alleviates pathological ultrastructural changes in Wolman disease cells. methyl-β cyclodextrin (MBCD), δ-tocopherol (δ-toco).

FIG. 7 is a set of photographs showing the electron microscopic analysis of Farber fibroblasts treated with methyl-β cyclodextrin (MBCD), α-cyclodextrin (alpha CD), or γ-cyclodextrin (gamma-CD).

FIG. 8 is a set of photographs showing the electron microscopic analysis of Tay-Sach, Fabry, and Farber fibroblasts treated with methyl-β cyclodextrin (MBCD).

FIG. 9 is a set of photographs showing the electron microscopic analysis of Wolman, NPA, Batten, and MSIIIB fibroblasts treated with methyl-β cyclodextrin (MBCD).

FIG. 10 is a set of photographs showing the electron microscopic analysis of Farber fibroblasts treated with δ-Tocopherol (DT) and methyl-β cyclodextrin (MBCD); δ-Tocopherol (DT) and α-cyclodextrin (a-T); δ-Tocopherol (DT) and γ-cyclodextrin (gamma-CD); δ-Tocopherol (DT) and Kleptose (also known as HBCD); α-Tocopherol (a-T) and methyl-β cyclodextrin (MBCD); and α-Tocopherol (a-T) and Kleptose. KLEPTOSE or TRAPPSOL are the brand names of the chemical HBCD or HBPCD.

FIGS. 11 (A and B) is a set of photographs that shows the effects of cyclodextrins and delta-tocopherol on reduction of cholesterol accumulation (Amplex-re cholesterol assay and filipin staining) and enlarged lysosomes (Lysotracker staining) in the NPC1 skin fibroblasts. Methyl-β cyclodextrin (MBCD), HBPCD (Kleptose).

FIGS. 12 (A and B) shows the effects of cyclodextrins and delta-tocopherol on reduction of cholesterol accumulation (Amplex-re cholesterol assay and filipin staining) and enlarged lysosomes (Lysotracker staining) in the NPC1 neuronal cells (NPC1-NSCs). (A) shows concentration-responses determined in Amplex-red cholesterol assay. (B) shows filipin and lysotracker staining. Methyl-β cyclodextrin (MBCD), δ-Tocopherol (δ-T)

FIGS. 13 (A and B) is a set of photographs that shows a comparison of the effect of single use of cyclodextrins with that in a combination with delta-tocopherol on reduction of cholesterol accumulation (filipin staining) and enlarged lysosomes ((Lysotracker staining) in NPC1 neuronal cells (NPC1-NSCs). (A) HBPCD+δ-Tocopherol. (B) MBCD+δ-Tocopherol.

FIG. 14 is a set of photographs showing lysotracker staining in 3123 treated with methyl-β cyclodextrin (MBCD); HBPCD or δ-Tocopherol (DT).

FIG. 15 is a set of photographs showing lysotracker staining in ML111 treated with methyl-β cyclodextrin (MBCD); HBPCD or δ-Tocopherol (DT).

FIG. 16 is a set of photographs showing lysotracker staining in MLIV treated with methyl-β cyclodextrin (MBCD); HBPCD or δ-Tocopherol (DT).

FIG. 17 is a set of photographs showing lysotracker staining in MPS1 treated with methyl-β cyclodextrin (MBCD); HBPCD or δ-Tocopherol (DT).

FIG. 18 is a set of photographs showing lysotracker staining in MPSV1 treated with methyl-β cyclodextrin (MBCD); HBPCD or δ-Tocopherol (DT).

DETAILED DESCRIPTION

Methods of Treatment

In one aspect, the invention provides a method of treating a lysosomal storage disorder in a subject, comprising determining that the subject is in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and other macromolecule accumulation, and administering to the subject in need thereof, an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another aspect, the invention provides a method of treating a lysosomal storage disorder in a subject, wherein the subject has been previously identified as being in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and other macromolecule accumulation, comprising administering to said subject in need thereof an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In one embodiment, the lysosomal storage disorder is treated by reducing the non-cholesterol lipid or non-cholesterol dominant lipid and other macromolecule accumulation in the subject.

In another embodiment, the non-cholesterol lipid is lipopigments, globotriaosylceramide, ceramide, sphingomyelin, heparan sulfate, partially degraded heparan sulfate, GM2 ganglioside, triglycerides, or cholesterol esters. The other macromolecules include proteins, glycoproteins (sugar containing proteins), mucopolysaccharides (long unbranched polysaccharides), and other cellular components.

A subject may be identified as having a lysosomal storage disease by presenting to a clinician with symptoms of a lysosomal storage disease, including but not limited to an enlarged liver and spleen. Storage may begin during early embryonic development, and the clinical presentation for lysosomal storage diseases can vary from an early and severe phenotype to late-onset mild disease. Said subject may be subject to a variety of diagnostic tests to determine if the subject has a lysosomal storage diease, and further to determine the presence of non-cholesterol lipids and macromolecules and non-cholesterol dominant lipids (i.e. non-cholesterol lipids that are present in an amount or percentage greater than that of cholesterol).

For example, ultrastructural examinations of skin biopsy specimens can be used to detect lysosomal accumulation of undegraded metabolites. A test of specific lysosomal enzyme activity can also be used to determine the presence of specific lysosomal enzymes. Moreover, correlation of both skin ultrastructure and assay for specific lysosomal enzymes in cultured dermal fibroblasts derived from the skin biopsy may also facilitate determination of cholesterol and non-cholesterol lipids, and diagnostic accuracy. Filipin staining is a well-known histochemical stain for cholesterol. Filipin is highly fluorescent and binds specifically to cholesterol. This method of detecting cholesterol in cell membranes is used clinically, for example in the study and diagnosis of Type C Niemann-Pick disease. Molecular genetic testing can be used, may be use to refine the enzymatic diagnosis. Other diagnostic methods to determine the present of cholesterol and non-cholesterol lipids include antibody immunostaining or mass spectrometry.

Lysosomal storage disorders include ˜40 to 50 inherited metabolic disorders caused by defects in lysosomal function. The incidence is about 1:5000-1:10,000 as a group of diseases. The term lysosomal refers to a recycling center in which cell membrane and other materials break down to small molecules for reuse. It has been found that deficiency of a single enzyme or proteins required for the metabolism or trafficking of lipids, glycoproteins and other macromolecules results in lipid accumulation in lysosome of cells. Excessive amount of lipids or other materials in lysosome causes enlargement of liver and spleen. Symptoms of neuronal degeneration are common clinical manifestations in patients with neuronal involvements.

Lysosomal storage disorders treated by the invention include, but are not limited to the following: Aspartylglucosaminuria, Wolman disease, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis, Gaucher disease, GM1-Gangliosidosis types I/II/III, GM2-Gangliosidosis, alpha-Mannosidosis types I/II, beta-Mannosidosis, Metachromatic leukodystrophy, Sialidosis types I/II, Mucolipidosis type IV, Scheie syndrome, Hunter syndrome, Sanfilippo syndrome A, Sanfilippo syndrome B, Sanfilippo syndrome C, Sanfilippo syndrome D, Galactosialidosis types I/II, Krabbe disease, Sandhoff disease, Vogt-Spielmeyer disease, Hurler syndrome, Niemann-Pick disease, I-cell disease (mucolipidosis II), pseudo-Hurler polydystrophy, Morquio syndrome, Maroteaux-Lamy syndrome, Sly syndrome, Mucopolysaccharidosis type IX, Multiple sulfatase deficiency, Batten disease, Tay-Sachs disease, Pompe disease, Batten disease, Batten disease, late infantile, Northern Epilepsy, Pycnodysostosis, Schindler disease, Sialuria, and Salla disease.

In certain embodiments, the lysosomal storage disorder is Tay-Sachs disease, Sphingolipidoses, Gaucher disease, Mucolipidosis, Galactosialidosis, Salla disorder, Cystinosis, Danon disease, Fabry disease, Farber disease, Lipofuscinoses, Pompe disease, Gangliodisosis, ISSD, Krabbe disease, leukodystrophy, Hurler disease, Scheie disease, Hunter disease, San Filippo disease, Sandhoff disease, Schinder disease, Batten disorder, or Wolman disease.

In a further embodiment, the lysosomal storage disorder is Tay-Sachs disease, Fabry disease, Farber disease, San Filippo disease, Batten disorder, or Wolman disease.

In certain embodiments, the cyclodextrin compound is of formula (I):

or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, wherein,

each R is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is optionally substituted; or —C(O)ORB, —OC(O)RB, —C(O)RB, or —C(O)NRARB;

each R1 is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, halogen, hydroxy, amino, —CN, —CF3, —N3, —NO2, —ORB, —SRB, —SORB, —SO2RB, —N(RB)S(O2)—RB, —N(RB) S(O2)NRARB, —NRARB, —C(O)ORB, —OC(O)RB, —C(O)RB, —C(O)NRARB, or —N(RB)C(O)RB; each of which is optionally substituted;

each RA is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is optionally substituted;

each RB is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is optionally substituted;

n is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and each m is independently 0, 1, 2, 3, 4, or 5.

In certain embodiments, each R is independently H, optionally substituted alkyl, —C(O)ORB, —OC(O)RB, —C(O)RB, or —C(O)NRARB.

In a further embodiment, each R is independently H, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, or octyl; wherein each is straight chain or branched.

In other embodiments, n is 1, 2, or 3.

In another embodiment, the cyclodextrin is 2-hydroxypropyl-β-cyclodextrin (2HPβCD), hydroxypropyl-β-cyclodextrin (HPβCD), methyl-β-cyclodextrin (MβCD), α-cyclodextrin, β-cyclodextrin, or γ-cyclodextrin, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In certain embodiments, the compound is:

TABLE 1 Types of beta-cyclodextrins # of Name sugars R # of R2 Name Heptakis(2,6-di-o- 7 a combination of R1 = —H in 14 MβCD methyl)-beta- position 3 and R2 = —Me in cyclodextrin positions 2 and 6 (2-Hydroxypropyl)- 7 a random combination of 1 to 10 Kleptose beta-cyclodextrin R1 = —H and R2 = —CH—CHOH—CH3 being 4 the most abundant species (2-Hydroxypropyl)- 7 a random combination of 4-10 Trappsol beta-cyclodextrin R1 = —H and R2 = —CH—CHOH—CH3 being 7 the most abundant species

In certain embodiments, the step of determining non-cholesterol lipid reduction or non-cholesterol dominant lipid and macromolecule accumulation in a subject comprises any one or more of the following.

Amplex-Red Cholesterol Assay—

Total cholesterol in patient cells was measured by the Amplex-Red Cholesterol Assay Kit (Invitrogen). The unesterified cholesterol was determined using the same kit without the enzyme acid lipase. Esterified cholesterol was determined as the difference between the total and unesterified cholesterol values. The cells were seeded to black, tissue culture-treated 96-well, 384-well or 1536-well plates at 4000, 1000, 300 cells/well in 100, 20 or 5 μl medium by a Multidrop Combi dispenser (Thermo Scientific, Waltham, Mass.) and cultured for 24 hr. The assay plates were added with compound dilution in DMSO solution using a Pintool station (Klaypsys, San Diego, Calif.) and cultured for 3 days. The cells were washed twice manually for 96-well or 384-well plates or using a centrifugation method in which the inverted plates were placed on a stack of paper towel and centrifuged at 800 rpm for 1 min followed by addition of 7 μl/well PBS (added gently with a 45 degree angled liquid dispenser (Klaypsys). The cholesterol assay mixture from the kit was added at 100, 20 or 2.5 μl/well for 96-well, 384-well or 1536-well plates and incubated for 1 hr at 37° C. The resulted fluorescence intensity was measured with excitation of 560 (±10) and emission of 590 (±10) in a fluorescence plate reader (Tecan, Durham, N.C.).

Nile-Red Staining—

The cells were cultured and treated as described above in 96-well plates. On the experimental day, cells were washed two times with PBS and live-stained with 1 uM Nile-red dye solution (prepared in cell culture medium) at 100 μl/well followed by an incubation at 37° C. for 10 min. After washed twice with PBS, the cells were fixed in 3.2% paraformaldehyde in PBS at 100 μl/well for 1 hr at RT. The nuclear staining was carried out by an addition of 100 μl/well 1 μg/ml Hoechst 33342 (Invitrogen) in PBS and incubation at RT for 30 min. The plate was washed twice with PBS and the images were measured in Incell2000 imaging plate reader with a FITC filter set (Ex=480±20 nm and Ex=525±36 nm) for the neutral lipids (cholesteryl esters and triglycerides) and a DAPI filter set for Hoechst nuclear staining.

LysoTracker Dye Staining—

The assay was optimized to visualize the enlarged lysosomes by applying appropriate concentration of LysoTracker dye in which the control cells exhibited minimal staining while the disease cells showed significant staining. The cells were cultured and treated as described above in 96-well plates. On the experimental day, cells were live-stained with 100 μl/well 50 nM LysoTracker-Red DND-99 dye (Invitrogen, # L-7528) in medium at 37° C. for 1 hr followed by plate washing twice with PBS. The plate was then fixed in 100 μl/well 3.2% formaldehyde for 1 hr and washed for two times with PBS. The nuclear staining were carried out by an addition of 100 μl/well 1 μg/ml Hoechst 33342 (Invitrogen) in PBS and incubation at RT for 30 min. After washing twice with PBS, the plates were stored at 4° C. until imaging analysis. DAPI filter set and TRITC filter set in the Incell2000 imaging plate reader were used to visualize Hoechst nuclear staining and LysoTracker staining, respectively.

Measurement of β-Hexosaminidase (HEXB) Release—

Fibroblasts were cultured in 24-well plates at 30,000 cells/well in 0.4 ml medium for one day at 37° C. After being washed twice with the assay buffer (DMEM with 2 mM D-mannose 6-phosphate sodium salt), the cells with 0.4 ml/well the assay buffer were incubated at 37° C. with 0.2 ml/well compound in assay buffer. At the 5, 10, 20, 30 and 40 min time points, 30 μl of assay buffer from each well in the 24-well plate were aliquoted into a 96-well black plate. The rest of assay buffer in the 24-well plate was discarded followed by addition of 0.6 ml 1% Triton-X100 in dH2O to lyse the cells. After incubation at 37° C. for 30 min, 6 μl/well cell lysate were added to the 96-well plate with 24 μl assay buffer followed by 90 μl/well 2.25 mM HEXB substrate, 4-Methylumbelliferyl N-acetyl-β-D-glucosaminide (Sigma-Aldrich, # M2133), in a 25 mM citric acid buffer at pH 4.5. The 96-well plate was then measured in the Tecan fluorescence plate reader (Ex=365±20 nm and Em=460±20 nm) after 1 hr incubation at 37° C. and addition of 100 μl/well stop solution (1 M glycine and 1 M NaOH at pH 10.5).

Intracellular and Lysosomal Ca2+ Measurements—

Intracellular cytosolic Ca2+ concentration was measured fluorescently using a Fluo-8 dye kit (ATT Bioquest, Sunnyvale, Calif.) as described previously. Briefly, fibroblasts were cultured at 2500 cell/well in 20 μl medium in black, clear bottom 384-well plates for 24 hr at 37° C. The calcium dye mixture was added at 20 μl/well and incubated at 37° C. for 30 min following by at RT for 30 min. The plates were then placed into a fluorescence kinetic plate reader (μCell, Hamamatsu, Hamamatsu City, Japan). The basal fluorescence intensity was recorded 10 times at 1 Hz for 10 seconds and the compound was then added at 20 μl/well inside the instrument followed by additional reading at 1 Hz for 5 min. The results were normalized to the average basal fluorescence intensity in ratio and the peak response (Max.) was used for the result calculation. The lysosomal Ca2+ induced by Gly-Phe β-naphthylamide (GPN) was measured similarly as that for cytosolic Ca2+ except 200 nM GPN was added instead of 8-T or α-T after the measurement of basal fluorescence intensity that released lysosomal Ca2+.

Electron Microscopy—

Fibroblast cells were seeded in 6-well plates at 150,000 cells/well in 5 ml medium and cultured for 1 day in the presence or absence of compounds. Cells were fixed in 2% glutaraldehyde, 0.1 M cacodylate buffer, pH 7.2 for 1 h at room temperature and then stored at 4° C. until TEM analysis was performed. The cells were post fixed in 1% osmium tetroxide in the same buffer for 1 hour and en bloc stained with 0.5% uranyl acetate in 0.1 M acetate buffer, pH 4.2. The cells were then dehydrated in graded ethanol solutions (35%, 50%, 70%, 95% and 100%) and infiltrated overnight in epoxy resin (Poly/Bed 812, Polysciences). After adding fresh pure resin the cell plates were cured for 72 h in 55° C. After removing the polystyrene plates, suitable areas for thin sectioning were selected, cut out with a jewelry saw and glued onto empty resin stubs. About 70 nm thin sections were cut on an ultramicrotome (Leica EM UC6) and mounted on naked copper grids. The thin sections were double stained (uranyl acetate and lead citrate), examined in a Hitachi H-7650 transmission electron microscope, and images were taken using an AMT CCD camera.

In various embodiments, the invention provides a method as described above further comprising the step of administering an additional therapeutic agent.

In certain embodiments, the additional therapeutic agent is a vitamin.

In a further embodiment, the additional therapeutic agent is vitamin E.

In other embodiments, the invention provides a method as described above wherein the step of administering the cyclodextrin comprises administering the compound orally, topically, parentally, intravenously or intramuscularly.

In certain embodiments, the invention provides a method comprising the step of administering an effective amount of the compound and a pharmaceutically suitable excipient.

In certain embodiments, the invention provides a method as described above wherein the subject is a human.

In various embodiments, the step of administering the cyclodextrin comprises administering the compound to a subject such as a human in a dosage of between about 0.01 μg/kg/day and 100 mg/kg/day, either in a single dose or per day.

In another aspect, the invention provides a method of treating a lysosomal storage disorder in a subject, comprising the step of administering to the subject an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and an additional therapeutic agent.

In certain embodiments, the additional therapeutic agent is a vitamin.

In a further embodiment, the additional therapeutic agent is vitamin E.

In certain aspects, the invention provides a method of reducing non-cholesterol lipids in a subject, the method comprising administering to the subject a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof; and detecting the amount of lipid reduction.

In one embodiment, the subject is identified as being in need of lipid reduction.

In another aspect, the invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention (any of the formulae presented herein), or a pharmaceutically acceptable salt, solvate or hydrate thereof, in combination with a pharmaceutically acceptable carrier or excipient.

In one embodiment, the pharmaceutical composition is in combination with a vitamin. In a further embodiment, the vitamin is vitamin E.

In another aspect, the invention provides a pharmaceutical composition comprising a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and vitamin E, together with a pharmaceutically-acceptable carrier or excipient.

In one embodiment, the cyclodextrin compound is of formula (I), or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another embodiment, the cyclodextrin compound is 2-hydroxypropyl-β-cyclodextrin (2HPβCD), hydroxypropyl-β-cyclodextrin (HPβCD), methyl-β-cyclodextrin (MβCD), α-cyclodextrin, β-cyclodextrin, or γ-cyclodextrin, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another aspect, the invention provides a method of treating a subject suffering from a lysosomal storage disorder, comprising the use of the pharmaceutical composition as described above, in combination with another agent.

In other aspects, the invention provides a kit comprising an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in unit dosage form, together with instructions for administering the compound to a subject suffering from a lysosomal storage disorder.

In one embodiment, the cyclodextrin compound is of formula (I), or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another embodiment, the cyclodextrin compound is 2-hydroxypropyl-β-cyclodextrin (2HPβCD), hydroxypropyl-β-cyclodextrin (HPβCD), methyl-β-cyclodextrin (MβCD), α-cyclodextrin, β-cyclodextrin, or γ-cyclodextrin, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

In another embodiment, the kit further comprises vitamin E.

In another embodiment, the invention provides a method as described above further comprising the step of synthesizing or obtaining the cyclodextrin compounds. Yet another embodiment of the present invention is a process of making any of the compounds delineated herein employing any of the synthetic means delineated herein, or using methods known to one of ordinary skill in the art.

In certain embodiments, cyclodextrins including α-, β- and γ-cyclodextrins increased intracellular Ca2+ and lysosomal exocytosis in both wild type and cells with LSDs (Wolman disease).

In various embodiments, cyclodextrins reduced enlarged lysosomes in six cell lines with LSDs.

In another embodiment, cyclodextrins reduced ultrastructural pathologic changes in cells with Wolman diseases and other cells.

In certain embodiments, cyclodextrins in combination with tocopherol synergistically/additively reduced cholesterol accumulation in cells of NPC and Wolman diseases.

An inhibitory amount or dose of the compounds of the present invention may range from about 0.1 mg/kg to about 500 mg/kg, alternatively from about 1 to about 50 mg/kg. Inhibitory amounts or doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents.

The term “inhibitory amount” of a compound of the present invention means a sufficient amount to decrease the disorder in a biological sample or a subject. It is understood that when said inhibitory amount of a compound of the present invention is administered to a subject it will be at a reasonable benefit/risk ratio applicable to any medical treatment as determined by a physician. The term “biological sample(s),” as used herein, means a substance of biological origin, which may be intended for administration to a subject. Examples of biological samples include, but are not limited to, blood and components thereof such as plasma, platelets, subpopulations of blood cells and the like; organs such as kidney, liver, heart, lung, and the like; sperm and ova; bone marrow and components thereof; or stem cells.

As referred to herein, the phrase “in combination with”, or “or conjunction with” when referring to administration of a cyclodextrin compound and an additional therapeutic agent (distinct from the cyclodextrin compound) such as a vitamin E compound is intended to refer to all forms of administration that provide the cyclodextrin and additional therapeutic compounds together, e.g. where the two compounds are administered concurrently (e.g. in a single unitary formulation) or sequentially in any order. For instance, in a suitable aspect, for sequential administration, the cyclodextrin compound and the additional therapeutic agent may be formulated separately and administered within about 0.25, 0.5, 1, 2, 5, 10, 15, 20, 30, 40, 50 or 60 minutes or more of each other. For sequential administration, preferably the cyclodextrin compound and the additional therapeutic agent may be formulated separately and administered within about 30, 20, 10 or 5 minutes or less of each other.

Upon improvement of a subject's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. The subject may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.

It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.

The total daily inhibitory dose of the compounds of this invention administered to a subject in single or in divided doses can be in amounts, for example, from 0.01 to 50 mg/kg body weight or more usually from 0.1 to 25 mg/kg body weight. Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose. In one embodiment, treatment regimens according to the present invention comprise administration to a patient in need of such treatment from about 10 mg to about 1000 mg of the compound(s) of this invention per day in single or multiple doses. In another embodiment, the treatment regimen comprises administration to a patient in need of such treatment from about 25 mg to about 6000 mg of a compound(s) of this invention per day in single or multiple doses. For instance a compound of the present invention can be administered to a patient twice a day with a total daily dose of 4000, 4200, 4400, 4600, 4800 or 5000 mg.

A preferred single use of cyclodextrin for mammals including humans is from 0.1 mg/Kg to 8 mg/Kg, more preferably 0.5 mg/kG or 1.0 mg/Kg to 2 mg/Kg, 3 mg/Kg, 4 mg/Kg, 5 mg/Kg, 6 mg/Kg, 7 mg/Kg or 84 mg/Kg. One specific preferred single use of cyclodextrin for mammals including humans is 3 mg/Kg.

In a combination therapy of a cyclodextrin compound administered together or otherwise in conjuction with a vitamin E compound such as delta-tocopherol, a preferred single dose for a mammal including a human may be from 0.05 to 1 mg/kg for each of the cyclodextrin compound and vitamin E compound (such as delta-tocopherol), more preferably 0.1 mg/Kg to 0.5 mg/Kg, 0.6 mg/Kg or 0.7 mg/Kg for each of a cyclodextrin compound and vitamin E compound such as delta-tocopherol, still more preferably 0.1 mg/Kg to 0.3 mg/Kg or 0.4 mg/Kg for each of a cyclodextrin compound and vitamin E compound such as delta-tocopherol.

In embodiments of the present invention, treatment of a lysosomal storage disorder with a combination of a cyclodextrin compound and vitamin E compound (such as delta-tocopherol) can allow for use of a lower dosage of the cyclodextrin compound to achieve a therapeutic effect than when the cyclodextrin compound is used alone. In certain aspects, the amount of the cyclodextrin compound administered is at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90%, less than an amount of the cyclodextrin compound necessary to achieve a therapeutic effect if administered without the vitamin E compound.

Biological Data

FIG. 1. (amplex red) Skin fibroblasts derived from NPC1 patients demonstrate profound and reproducible cholesterol accumulation in late endosomes and lysosomes and, therefore, provide a robust cellular model of NPC1 disease. Using a phenotypic screen with a biochemical assay (Amplex Red) to measure unesterified cholesterol; Delta-Tocopherol (“δ-T”; or “Delta-T”) δ-T was identified as a lead compound that dramatically reduces cellular cholesterol accumulation in a concentration dependent manner. We further evaluated effect of cyclodextrins alone and in combination with delta Tocopherol in other lysosomal storage disorders and we found that alpha-CD, beta-CD, and gamma-CD can reduce cholesterol accumulation, and MBCD was most potent.

FIG. 2. (Nile Red) Cells were cultured in the presence of delta tocopherol, alpha tocopherol and MBCD plus in combination for 3 days and then stained for neutral lipid with Nile red. Delta Tocopherol and MBCD treatment reduces accumulation of neutral lipids and it is more pronounced when used in combination. Alpha Tocopherol was not as potent.

FIG. 3. (Hex assay) Delta-T stimulates lysosomal exocytosis in Wolman fibroblasts. 2-hydroxypropyl-beta-cyclodextrin has been reported to promote a calcium-dependent lysosomal exocytosis, which offers a potential mechanism for its cholesterol-reducing effect in LSD fibroblast. We measured lysosomal exocytosis in delta-T-treated Wolman fibroblasts by determining the activity of beta-hexosaminidase (HEXB), a lysosomal enzyme, in the extracellular medium. We found that HEXB activity increased in culture medium after 40 uM δ-Tocopherol treatment for 24 hours compared with the vehicle treated cells. These results demonstrate that the pharmacological effect of delta-T may be mediated by the increase of cytosolic Ca2+ and enhancement of lysosomal exocytosis.

FIG. 4. (calcium assay) Delta-T increases intracellular Ca2+ concentration and ameliorates lysosomal calcium deficiency in NPC1 cells—Increase in the concentration of intracellular Ca2+, an important second messenger, triggers a variety of cellular responses including lysosomal exocytosis. In NPC1 fibroblasts there is a dysregulation of calcium homeostasis, as evidenced by lysosomal Ca2+ deficiency. Treatments that compensate for loss of lysosomal Ca2+ (e.g., curcumin) have been reported to reduce cholesterol storage in NPC1 cells. To explore whether delta-T may similarly exert its effects through changes in intracellular Ca2++, we measured cytosolic calcium levels in both NPC1 and Wolman cells following the treatment with δ-T. We found that δ-T stimulated a transient increase of cytosolic Ca2+ in both NPC1 and Wolman fibroblasts, as well as in control fibroblasts. In addition, the intracellular Ca2+ response to delta-T was independent of extracellular Ca2+ concentration, indicating that Ca2+ was released from intracellular storage sites such as the ER in response to δ-T. We further studied the effect of delta-T on lysosomal Ca2+ released by Gly-Phe β-naphthylamide (GPN) in NPC1 fibroblasts. Consistent with an earlier report, lysosomal Ca2+ was reduced in NPC1 cells compared with that in control cells. Treatment of NPC1 fibroblasts with 40 uM delta-T for 24 hours significantly increased lysosomal Ca2+.

FIG. 5. Based on the data for both NPC1 and Wolman cells, we hypothesized that the pharmacological effect of delta-T on the intracellular Ca2+ and lysosomal exocytosis is a general mechanism for elimination of lysosomal storage. To test this hypothesis we measured the ability of delta-T to decrease acidic/lysosomal compartment size as determined by LysoTracker staining in fibroblasts derived from patients with six other diseases. Lysosomal storage occurs in these fibroblasts consists of ceroid/lipofuscin in Batten (CLN2), globotriaosylceramide in Fabry, ceramide in Farber, sphingomyelin in NPA, partially degraded heparan sulfate in Sanfilippo type B, and GM2 ganglioside in Tay-Sachs (Table S1). Whereas untreated fibroblasts showed increased LysoTracker staining, indicating the enlarged lysosomes, treatment with 40 uM delta-T significantly reduced the LysoTracker staining in all six fibroblast cell lines studied. Thus, the amelioration of lysosomal pathology by delta-T, initially demonstrated in NPC1 and Wolman cells, can be generalized to other lysosomal storage diseases.

The mixed lipid storage phenotype results in a marked enlargement of lysosomes in the NPC1 and Wolman fibroblasts. Therefore, we next determined whether the enlarged lysosomes in these cells could be reduced by the treatment with δ-T. LysoTracker, a probe which stains the intracellular acidic compartment, has been used to visualize the enlarged endolysosomal compartment in NPC1 cells. We found increased LysoTracker staining in both NPC1 and Wolman fibroblasts, as expected. Treatment with either 40 uM delta-T or 300 uM MBCD significantly reduced LysoTracker staining in both types of fibroblasts.

FIG. 6. Both NPC1 and Wolman fibroblasts have a distinct ultrastructural phenotype that is evident by electron microscopy. The reduction of acidic cellular compartments by delta-T treatment is consistent with decreased intracellular storage that was confirmed by alleviation of the ultrastructural pathology.

The electron microscopic images exhibited enlarged lysosomes full of lamellated membranes and dense osmiophilic material in NPC1 cells and lipid droplet-like and cleft-like lysosomes in Wolman cells. Treatment with 40 uM delta-T significantly reduced the characteristic storage materials in lysosomes of both cell types. Together, these findings demonstrate that the delta-T-mediated cholesterol reduction is associated with alleviation of the disease phenotypes in NPC1 and Wolman cells.

We have found that alpha-CD, beta-CD, and gamma-CD can reduce cholesterol accumulation in NPC cells. We also found that these CDs increased intracellular Ca2+ and enhanced exocytosis. The ranking order of cholesterol reduction effect is MBCD>alpha-CD>gamma-CD. In addition we found that the CD treatment reduced the pathological changes in the ultrastructure of NPC cells using the electron microscopy analysis. We also found that CDs reduced enlarged lysosomes in the primary fibroblasts of Wolman disease another lysosomal storage disease that exhibits cholesterol ester accumulation due to the malfunction of acid lipase in lysosome. The electron microscopy data indicated that the CD effect is more significant than that of delta-tocopherol in the Wolman cells.

We also found the synergy between CD and delta-tocopherol on the NPC cells and other 6 lysosomal storage disease cells including Wolman, Niemann Pick Type A, Farber, Tay-Sachs, MSIIIB and CLN2 (Batten) diseases. The fluorescence tagged CD study indicated that CD enters cell and comes out of cell quickly, indicating via exocytosis. We also have the data demonstrate that alpha-CD, beta-CD and gamma-CD enhance exocytosis.

DEFINITIONS

Listed below are definitions of various terms used to describe this invention. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group. The number of carbon atoms in a hydrocarbyl substituent can be indicated by the prefix “Cx-Cy,” where x is the minimum and y is the maximum number of carbon atoms in the substituent. Likewise, a Cx chain means a hydrocarbyl chain containing x carbon atoms.

The prefix “halo” indicates that the substituent to which the prefix is attached is substituted with one or more independently selected halogen radicals. For example, “C1-C6haloalkyl” means a C1-C6alkyl substituent wherein at least one hydrogen radical is replaced with a halogen radical.

If a linking element in a depicted structure is “absent” or “a bond”, then the left element in the depicted structure is directly linked to the right element in the depicted structure. For example, if a chemical structure is depicted as X-(L)n-Y wherein L is absent or n is 0, then the chemical structure is X—Y.

The term “alkyl” as used herein, refers to a saturated, straight- or branched-chain hydrocarbon radical. For example, “C1-C8 alkyl” contains from one to eight carbon atoms. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl, heptyl, octyl radicals and the like.

The term “alkenyl” as used herein, denotes a straight- or branched-chain hydrocarbon radical containing one or more double bonds. For example, “C2-C8 alkenyl” contains from two to eight carbon atoms. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, heptenyl, octenyl and the like.

The term “alkynyl” as used herein, denotes a straight- or branched-chain hydrocarbon radical containing one or more triple bonds. For example, “C2-C8 alkynyl” contains from from two to eight carbon atoms. Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl and the like.

The term “cycloalkyl” denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound. Examples of cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[2.2.1]heptyl, and bicyclo[2.2.2]octyl and the like. The terms “carbocycle” or “carbocyclic” or “carbocyclyl” refer to a saturated (e.g., “cycloalkyl”), partially saturated (e.g., “cycloalkenyl” or “cycloalkynyl”) or completely unsaturated (e.g., “aryl”) ring system containing zero heteroatom ring atom. A carbocyclyl may be, without limitation, a single ring, or two or more fused rings, or bridged or spiro rings. A carbocyclyl may contain, for example from 3 to 10 ring members (i.e., C3-C10carbocyclyl, such as C3-C10cycloalkyl). A substituted carbocyclyl may have either cis or trans geometry. Representative examples of carbocyclyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopentenyl, cyclopentadienyl, cyclohexadienyl, adamantyl, decahydro-naphthalenyl, octahydro-indenyl, cyclohexenyl, phenyl, naphthyl, fluorenyl, indanyl, 1,2,3,4-tetrahydro-naphthyl, indenyl, isoindenyl, bicyclodecanyl, anthracenyl, phenanthrene, benzonaphthenyl (also known as “phenalenyl”), decalinyl, and norpinanyl and the like. A carbocyclyl group can be attached to the parent molecular moiety through any substitutable carbon atom of the group.

The term “aryl” refers to an aromatic carbocyclyl containing from 6 to 14 carbon ring atoms. Non-limiting examples of aryls include phenyl, naphthalenyl, anthracenyl, and indenyl and the like. An aryl group can be connected to the parent molecular moiety through any substitutable carbon atom of the group.

The term “heteroaryl” means an aromatic heterocyclyl typically containing from 5 to 18 ring atoms, wherein at least one ring atom is a heteroatom. A heteroaryl may be a single ring, or two or more fused rings. Non-limiting examples of five-membered heteroaryls include imidazolyl; furanyl; thiophenyl (or thienyl or thiofuranyl); pyrazolyl; oxazolyl; isoxazolyl; thiazolyl; 1,2,3-, 1,2,4-, 1,2,5-, and 1,3,4-oxadiazolyl; and isothiazolyl. Non-limiting examples of six-membered heteroaryls include pyridinyl; pyrazinyl; pyrimidinyl; pyridazinyl; and 1,3,5-, 1,2,4-, and 1,2,3-triazinyl. Non-limiting examples of 6/5-membered fused ring heteroaryls include benzothiofuranyl, isobenzothiofuranyl, benzisoxazolyl, benzoxazolyl, purinyl, and anthranilyl. Non-limiting examples of 6/6-membered fused ring heteroaryls include quinolinyl; isoquinolinyl; and benzoxazinyl (including cinnolinyl and quinazolinyl).

The term “heterocycloalkyl” refers to a non-aromatic 3-, 4-, 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused system, where at least one of the ring atoms is a heteroatom, and where (i) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (ii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above rings may be fused to a benzene ring. Representative heterocycloalkyl groups include, but are not limited to, [1,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl and the like.

The terms “heterocyclic” or “heterocycle” or “heterocyclyl” refer to a saturated (e.g., “heterocycloalkyl”), partially unsaturated (e.g., “heterocycloalkenyl” or “heterocycloalkynyl”) or completely unsaturated (e.g., “heteroaryl”) ring system, where at least one of the ring atoms is a heteroatom (i.e., nitrogen, oxygen or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur. A heterocyclyl group can be linked to the parent molecular moiety via any substitutable carbon or nitrogen atom in the group, provided that a stable molecule results. A heterocyclyl may be, without limitation, a single ring. Non-limiting examples of single-ring heterocyclyls include furanyl, dihydrofuranyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiodiazolyl, oxathiazolyl, oxadiazoly, pyranyl, dihydropyranyl, pyridinyl, piperidinyl, pyridazinyl, pyrimidinyl, pyrazinyl, piperazinyl, triazinyl, isoxazinyl, oxazolidinyl, isoxazolidinyl, oxathiazinyl, oxadiazinyl, morpholinyl, azepinyl, oxepinyl, thiepinyl, or diazepinyl. A heterocyclyl may also include, without limitation, two or more rings fused together, such as, for example, naphthyridinyl, thiazolpyrimidinyl, thienopyrimidinyl, pyrimidopyrimidinyl, or pyridopyrimidinyl. A heterocyclyl may comprise one or more sulfur atoms as ring members; and in some cases, the sulfur atom(s) is oxidized to SO or SO2. The nitrogen heteroatom(s) in a heterocyclyl may or may not be quaternized, and may or may not be oxidized to N-oxide. In addition, the nitrogen heteroatom(s) may or may not be N-protected.

The terms “optionally substituted”, “optionally substituted alkyl,” “optionally substituted “optionally substituted alkenyl,” “optionally substituted alkynyl”, “optionally substituted carbocyclic,” “optionally substituted aryl”, “optionally substituted heteroaryl,” “optionally substituted heterocyclic,” and any other optionally substituted group as used herein, refer to groups that are substituted or unsubstituted by independent replacement of one, two, or three or more of the hydrogen atoms thereon with typical substituents including, but not limited to:

-alkyl, -alkenyl, -alkynyl, -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, -heterocycloalkyl, -cycloalkyl, -carbocyclic, -heterocyclic,

—F, —Cl, —Br, —I,

—OH, protected hydroxy, alkoxy, oxo, thiooxo,

—NO2, —CN, CF3, N3,

—NH2, protected amino, —NH alkyl, —NH alkenyl, —NH alkynyl, —NH cycloalkyl, —NH— aryl, —NH— heteroaryl, —NH— heterocyclic, -dialkylamino, -diarylamino, -diheteroarylamino,

—O— alkyl, —O— alkenyl, —O— alkynyl, —O— cycloalkyl, —O-aryl, —O-heteroaryl, —O— heterocyclic,

—C(O)— alkyl, —C(O)— alkenyl, —C(O)— alkynyl, —C(O)— cycloalkyl, —C(O)-aryl, —C(O)— heteroaryl, —C(O)-heterocycloalkyl,

—CONH2, —CONH— alkyl, —CONH— alkenyl, —CONH— alkynyl, —CONH— cycloalkyl, —CONH-aryl, —CONH-heteroaryl, —CONH-heterocycloalkyl,

—OCO2— alkyl, —OCO2— alkenyl, —OCO2— alkynyl, —OCO2— cycloalkyl, —OCO2-aryl, —OCO2-heteroaryl, —OCO2-heterocycloalkyl, —OCONH2, —OCONH— alkyl, —OCONH— alkenyl, —OCONH— alkynyl, —OCONH— cycloalkyl, —OCONH— aryl, —OCONH— heteroaryl, —OCONH— heterocycloalkyl,

—NHC(O)— alkyl, —NHC(O)— alkenyl, —NHC(O)— alkynyl, —NHC(O)— cycloalkyl, —NHC(O)-aryl, —NHC(O)-heteroaryl, —NHC(O)-heterocycloalkyl, —NHCO2— alkyl, —NHCO2-alkenyl, —NHCO2— alkynyl, —NHCO2-cycloalkyl, —NHCO2— aryl, —NHCO2— heteroaryl, —NHCO2— heterocycloalkyl, —NHC(O)NH2, —NHC(O)NH— alkyl, —NHC(O)NH— alkenyl, —NHC(O)NH— alkenyl, —NHC(O)NH— cycloalkyl, —NHC(O)NH-aryl, —NHC(O)NH-heteroaryl, —NHC(O)NH-heterocycloalkyl, NHC(S)NH2, —NHC(S)NH— alkyl, —NHC(S)NH— alkenyl, —NHC(S)NH— alkynyl, —NHC(S)NH— cycloalkyl, —NHC(S)NH-aryl, —NHC(S)NH-heteroaryl, —NHC(S)NH-heterocycloalkyl, —NHC(NH)NH2, —NHC(NH)NH— alkyl, —NHC(NH)NH-alkenyl, —NHC(NH)NH— alkenyl, —NHC(NH)NH— cycloalkyl, —NHC(NH)NH-aryl, —NHC(NH)NH-heteroaryl, —NHC(NH)NH-heterocycloalkyl, —NHC(NH)— alkyl, —NHC(NH)— alkenyl, —NHC(NH)— alkenyl, —NHC(NH)— cycloalkyl, —NHC(NH)-aryl, —NHC(NH)— heteroaryl, —NHC(NH)-heterocycloalkyl,

—C(NH)NH— alkyl, —C(NH)NH— alkenyl, —C(NH)NH— alkynyl, —C(NH)NH— cycloalkyl, —C(NH)NH-aryl, —C(NH)NH-heteroaryl, —C(NH)NH-heterocycloalkyl,

—S(O)— alkyl, —S(O)— alkenyl, —S(O)— alkynyl, —S(O)— cycloalkyl, —S(O)-aryl, —S(O)-heteroaryl, —S(O)-heterocycloalkyl —SO2NH2, —SO2NH— alkyl, —SO2NH— alkenyl, —SO2NH-alkynyl, —SO2NH— cycloalkyl, —SO2NH— aryl, —SO2NH— heteroaryl, —SO2NH— heterocycloalkyl,

—NHSO2— alkyl, —NHSO2— alkenyl, —NHSO2— alkynyl, —NHSO2— cycloalkyl, —NHSO2-aryl, —NHSO2-heteroaryl, —NHSO2-heterocycloalkyl,

—CH2NH2, —CH2SO2CH3, polyalkoxyalkyl, polyalkoxy, -methoxymethoxy, -methoxyethoxy, —SH, —S— alkyl, —S— alkenyl, —S— alkynyl, —S— cycloalkyl, —S-aryl, —S— heteroaryl, —S-heterocycloalkyl, or methylthiomethyl.

It is understood that the aryls, heteroaryls, carbocycles, heterocycles, alkyls, and the like can be further substituted.

The terms “halo” and “halogen,” as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.

The term “subject” as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be either a patient or a healthy human.

The term “non-cholesterol lipid” is meant to refer to any lipid that is not cholesterol, for example a macromolecule. Exemplary non-cholestorl lipids include, but are not limited to, lipopigments, globotriaosylceramide, ceramide, sphingomyelin, heparan sulfate, partially degraded heparan sulfate, GM2 ganglioside, triglycerides, and cholesterol esters, and derivatives thereof. A “non-cholesterol dominant lipid” is meant to refer to any lipid that is not cholesterol, that is present in an amount greater than cholesterol making it the non-cholesterol dominant lipid.

The term “leaving group,” or “LG”, as used herein, refers to any group that leaves in the course of a chemical reaction involving the group and includes but is not limited to halogen, brosylate, mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate groups, for example.

The term “protected hydroxy,” as used herein, refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including benzoyl, acetyl, trimethylsilyl, triethylsilyl, methoxymethyl groups, for example.

The term “hydroxy protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect a hydroxy group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the hydroxy protecting group as described herein may be selectively removed. Hydroxy protecting groups as known in the are described generally in T. H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of hydroxy protecting groups include benzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4-bromobenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, methoxycarbonyl, tert-butoxycarbonyl, isopropoxycarbonyl, diphenylmethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, 2-(trimethylsilyl)ethoxycarbonyl, 2-furfuryloxycarbonyl, allyloxycarbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t-butyl, 2,2,2-trichloroethyl, 2-trimethylsilyl ethyl, 1,1-dimethyl-2-propenyl, 3-methyl-3-butenyl, allyl, benzyl, para-methoxybenzyldiphenylmethyl, triphenylmethyl(trityl), tetrahydrofuryl, methoxymethyl, methylthiomethyl, benzyloxymethyl, 2,2,2-triehloroethoxymethyl, 2-(trimethylsilyl)ethoxymethyl, methanesulfonyl, para-toluenesulfonyl, trimethylsilyl, triethylsilyl, triisopropylsilyl, and the like. Preferred hydroxy protecting groups for the present invention are acetyl (Ac or —C(O)CH3), benzoyl (Bz or —C(O)C6H5), and trimethylsilyl (TMS or —Si(CH3)3).

The term “amino protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect an amino group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the amino protecting group as described herein may be selectively removed Amino protecting groups as known in the are described generally in T. H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of amino protecting groups include, but are not limited to, t-butoxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyloxycarbonyl, and the like.

The term “protected amino,” as used herein, refers to an amino group protected with an amino protecting group as defined above.

The term “alkylamino” refers to a group having the structure —N(RaRb), where Ra and Rb are independent H or alkyl.

As used herein, the term “pharmaceutically acceptable salt” refers to those salts of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid. Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts, or salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, or magnesium salts, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate.

As used herein, the term “pharmaceutically acceptable ester” refers to esters of the compounds formed by the process of the present invention which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.

The term “pharmaceutically acceptable prodrugs” as used herein refers to those prodrugs of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the present invention. “Prodrug”, as used herein means a compound which is convertible in vivo by metabolic means (e.g. by hydrolysis) to afford any compound delineated by the formulae of the instant invention. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). “Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 8:1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer, “Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzymology,” John Wiley and Sons, Ltd. (2002).

This invention also encompasses pharmaceutical compositions containing pharmaceutically acceptable prodrugs of compounds of the invention. For example, compounds of the invention having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of compounds of the invention. The amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Additional types of prodrugs are also encompassed. For instance, free carboxyl groups can be derivatized as amides or alkyl esters. Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxy carbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 1 15. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers wherein the acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities.

As used herein, “solvate” refers to the physical association of a compound of the invention with one or more solvent molecule, whether organic or inorganic. This physical association often includes hydrogen bonding. In certain instances, the solvate is capable of isolation, for example, when one or more solvate molecules are incorporated in the crystal lattice of the crystalline solid.

Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term “stable”, as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).

Pharmaceutical Compositions

The pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term “pharmaceutically acceptable carrier” means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The pharmaceutical compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, or as an oral or nasal spray.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water, alcohol or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, polysorbate, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), mono- or diglycerides, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, antioxidants, sweetening, flavoring, and perfuming agents. The liquid dosage form can also be encapsulated in a gelatin capsule, wherein a compound of the present invention can be dissolved in a pharmaceutically acceptable carrier containing, for example, one or more solubilizating agents (e.g., polysorbate 80 and mono and diglycerides), and other suitable excipients (e.g., an antioxidants such as ascorbyl palmitate, or a sweetening or flavoring agent).

Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.

In certain preferred embodiments, the compositions of the present invention are administered intracranially, for instance injected into the brain, such as by direct injection into the brain. Direct injection may be performed by intraventricular and intracerebral routes. Injection of the compositions into the brain can also be performed using a device for administration. Because cyclodextrin and delta-tocopherol may pose a challenge with brain penetration and quick drug metabolism, direct administration of the drugs into the central nervous system may be achieved by using epidural (injection or infusion into the epidural space), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), or intrathecal (into the spinal canal) injection. Pathan et al. (Recent Patents on Drug Delivery & Formulation 2009, 3, 71-89), incorporated by reference in its entirety herein, describes some method of administration of a composition to the brain.

In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Immediate release forms are also contemplated by the present invention.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.

Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

The active compounds can also be in micro-encapsulated form with one or more excipients as noted above.

The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.

Preferably, a compound of the invention is formulated in a solid dispersion, where the compound can be molecularly dispersed in a matrix which comprises a pharmaceutically acceptable, hydrophilic polymer. The matrix may also contain a pharmaceutically acceptable surfactant. Suitable solid dispersion technology for formulating a compound of the invention includes, but is not limited to, melt extrusion, spray drying, or solvent evaporization.

Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.

The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to the compounds of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.

Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

The compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-, or as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optical isomers may be prepared from their respective optically active precursors by the procedures described above, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et al., Enantiomers, Racemates, and Resolutions (John Wiley & Sons, 1981). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon-carbon double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion.

The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. As can be appreciated by the skilled artisan, further methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and one of ordinary skill in the art will recognize that variation of the reaction conditions can produce the desired bridged macrocyclic products of the present invention. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof.

The compounds of this invention may be modified by appending various functionalities via any synthetic means delineated herein to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.

The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.

EXAMPLES

The compounds and processes of the present invention will be better understood in connection with the following examples, which are intended as an illustration only and not to limit the scope of the invention. The following examples can be prepared according to the schemes as described above, or according to the synthetic steps as described below. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, formulations and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims.

The chemical structures herein contain certain —NH—, —NH2 (amino) and —OH (hydroxyl) groups where the corresponding hydrogen atom(s) may not explicitly appear; however they are to be read as —NH—, —NH2 or —OH as the case may be.

Example 1 δ-Tocopherol and Cyclodextrin Treatment Reduces Lipid Accumulation in Lysosomes of Lysosome Storage Disorder Cells

The effect of methyl-β-cyclodextrin on lipid accumulation in fibroblast lines derived from patients with Wolman disease was investigated. Wolman fibroblasts were treated with δ-tocopherol, α-tocopherol, methyl-β-cyclodextrin, or combinations of δ-tocopherol and methyl-β-cyclodextrin or α-tocopherol and methyl-β-cyclodextrin. Total cholesterol and free cholesterol were then measured using the Amplex-Red Cholesterol Oxidase assay (Invitrogen) according to the manufacturer's instructions. As shown in FIGS. 1A-1C (Total cholesterol) and in FIGS. 1D-1F (Free cholesterol), treatment with δ-tocopherol, methyl-β-cyclodextrin, or combinations of δ-tocopherol and methyl-β-cyclodextrin or α-tocopherol and methyl-β-cyclodextrin caused a significant reduction in total cholesterol and free cholesterol in Wolman fibroblasts. To further characterize the effect of treatment on lipid accumulation, treated fibroblasts were evaluated using a Nile Red assay to measure neutral lipid accumulation. In brief, cells treated with various drugs were stained with Nile Red which selectively labels lipid accumulations within cells. The Nile Red staining was visualized by fluorescent microscopy. As shown in FIG. 2, untreated Wolman fibroblasts show cytoplasmic droplets of neutral lipid accumulation. These neutral lipid accumulations were significantly reduced upon treatment with δ-tocopherol or methyl-β-cyclodextrin. Interestingly, α-tocopherol treatment failed to show any effect on neutral lipid accumulation.

The effects of δ-tocopherol, α-tocopherol, methyl-β-cyclodextrin, or combinations of δ-tocopherol and methyl-β-cyclodextrin or α-tocopherol and methyl-β-cyclodextrin treatment on lysosomal exocytosis were determined using the HEXB assay. In brief, the level of lysosomal exocytosis was determined by measuring the level of the lysosomal enzyme HEXB secreted into the culture medium following treatment with drug. As shown in FIG. 3, treatment with α-tocopherol, methyl-β-cyclodextrin, or combinations of δ-tocopherol and methyl-β-cyclodextrin or α-tocopherol and methyl-β-cyclodextrin resulted in significant increases in lysosomal exocytosis as determined by HEXB secretions. The highest levels of exocytosis were seen in Wolman fibroblasts treated with the combination of δ-tocopherol and methyl-β-cyclodextrin.

To further characterize the effects of cyclodextrin on lysosome function in fibroblast lines derived from patients with lysosomal storage diseases (Wolman, Tay-Sach, Farber, Battern, and Fabry), the effect of cycoldextrin treatment on lysosomal Ca2+ release was determined. Both wild-type and lysosomal storage disease fibroblasts were treated with combinations of δ-tocopherol and methyl-β-cyclodextrin or α-tocopherol and methyl-β-cyclodextrin and the levels of lysosomal Ca2+ release stimulated by 200 nM Gly-Phe β-naphthylamide (GPN) was measured. As shown in FIG. 4, untreated lysosomal storage disease fibroblasts displayed reduced Ca2+ release compared to untreated wild-type fibroblasts. However, treatment of Wolman fibroblasts, Tay-Sach fibroblasts, Farber fibroblasts, Battern fibroblasts, and Fabry fibroblasts with combinations of δ-tocopherol and methyl-β-cyclodextrin or α-tocopherol and methyl-β-cyclodextrin restored lysosomal Ca2+ release to those levels seen in wild-type fibroblasts.

The effect of various cyclodextrins, δ-tocopherol, and combinations of cyclodextrins and δ-tocopherol on lipid accumulation and lysosome size in wild-type and lysosomal storage disease fibroblasts (Wolman, Tay-Sach, Fabry, Farber, and MPSIIIB fibroblasts) was determined using the Lysotracker assay. In brief, cells were treated with various combinations and concentrations of drugs followed by staining with the Lyso tracker dye which is a basophilic fluoresecent probe that accumulates in acidic compartments, i.e. lysosomes, within the cell. As shown in FIGS. 5A-5D, Lysotracker staining revealed enlarged lysosomes in Wolman, Tay-Sach, Fabry, Farber, and MPSIIIB fibroblasts compared to wild-type cells. Treatment with δ-tocopherol or methyl-β-cyclodextrin alone reduced lipid accumulation and lysosome size whereas treatment with other cyclodextrins alone did not have a profound effect. Moreover, the combination of δ-tocopherol and methyl-β-cyclodextrin resulted in a significant reduction in lipid accumulation and lysosome size.

Example 2 δ-Tocopherol and Cyclodextrin Treatment Resulted in Alleviation of the Pathological Ultrastructural Changes in Lysosomes of Cells with Lysosome Storage Disorders

The effects of δ-tocopherol and methyl-β-cyclodextrin on the ultrastructural pathology of lysosomes in lysosome storage disorder cells were investigated by electron microscopy. In brief, following treatment of wild-type and lysosome storage disorder fibroblasts with δ-tocopherol, methyl-β-cyclodextrin, or δ-tocopherol and methyl-β-cyclodextrin, the fibroblasts were fixed and embedded. Thin sections of the embedded cells were then prepared and the ultrastructural pathology was examined by electron microscopy. As shown in FIG. 6, untreated Wolman fibroblasts have lamellated and osmophilic structures within the lysosomes. In addition, the cells also have the typical elongated and cleft-shaped lipid droplets in the lysomes. However, these abnormal structures were significantly reduced by treatment with δ-tocopherol and/or methyl-β-cyclodextrin. The effects of δ-tocopherol and/or methyl-β-cyclodextrin on the ultrastructural pathology of other lysosome disorder fibroblasts—Farber (FIGS. 7, 8, and 10); Tay-Sachs (FIG. 8); Fabry (FIG. 8); Wolman (FIG. 9); NPA (FIG. 9); Batten (FIG. 9); MSIIIB (FIG. 9)—were analyzed by electron microscopy. As shown in FIGS. 7-10, treated cells appear as typical fibroblasts (elongated shape, well developed nuclei, normal mitochondria, swollen smooth and rough ER) but with significant amounts of endosomal (in particular multivesicular bodies) and lysosomal compartments filled with lipid droplets and multilamellar bodies. These compartments are typical for Farber cells as seen before. Most cells have only small areas of these structures but a few cells are very much filled with these structures.

Example 3 Effect of Cyclodextrin in Single Use and in Combination with δ-Tocopherol in Human NPC1 Fibroblasts and Neural Stem Cells (NPC-NSCs)

In another set of experiments, in skin fibroblasts derived from NPC1 patients, high concentrations of HBPCD (in millimolars) is needed for reduction of cholesterol accumulation and enlarged lysosomes (FIG. 11A). However, the small concentration of 50 uM of HBPCD in combination with 10 uM delta-tocopherol reached the same effect as 5 mM HBPCD. Although 160 uM MBCD almost completely reversed the phenotype of NPC1 cells, a much smaller concentration of 20 uM MBCD in combination with 10 uM delta-tocopherol reached the similar results as those obtained with higher concentration of MBCD used along (FIG. 11B). Together, the data indicate that MBCD is more potent (over 30 fold) than HBPCD for reduction of cholesterol accumulation and enlarged lysosome size in NPC1 fibroblasts. In the combination with 10 uM delta-tocopherol, much smaller concentrations of HBPCD and MBCD are needed compared to those when both drugs used along.

Example 4 Effect of Cyclodextrin in Single Use and in Combination with δ-Tocopherol in Neural Cells Derived from NPC1 Patients

Since major symptoms of NPC disease are within the central nervous system, the human NPC1 neuronal cells are better representative as a NPC disease model for drug evaluation. Induced pluripotent stem cells (IPSCs) from the NPC1 skin fibroblasts were generated and differentiated into neural stem cells (NPC1-NSCs). In the Amplex-red cholesterol assay, the IC50 values of HBPCD and MBCD were 12 and 10 uM, respectively in the NPC1-NSCs, while the IC50 for delta-tocopherol was 18 uM (FIG. 12A). In the fluorescence microscopy experiments, the effects of HBPCD and MBCD on reduction of cholesterol accumulation (filipin staining) and enlarged lysosomes (Lysotracker staining) were also better than those of delta-tocopherol (FIG. 12B). Taken together, the data indicate that HBPCD is much more potent in the human NPC1 neuronal cells than that in the NPC1 skin fibroblasts, whereas the potency of delta-tocopherol is weaker in NPC1 neuronal cells compared to that in the NPC1 fibroblasts.

Example 5 Combination Therapy of Cyclodextrin and δ-Tocopherol Effectively Reduced the Concentrations of Individual Compounds and Increased the Effect on Reduction of Cholesterol Accumulation and Enlarged Lysosomes in NPC1-NSCs

A much reduced concentration of 50 uM HBPCD in combination with 10 uM delta-tocopherol was determined to reach the same effect of 5 mM HBPCD used alone on reduction of cholesterol accumulation and enlarged lysosomes in the NPC1-NSCs (FIG. 13A). Similarly, the effect of 20 uM MBCD in combination with 10 uM delta-tocopherol was similar as 160 uM MBCD used alone in the NPC1-NSCs (FIG. 13B). The data demonstrate that the combination therapy of lower concentration of cyclodextrin and delta-tocopherol could achieve the similar therapeutic effect on reduction of cholesterol accumulation and enlarged lysosomes in the NPC1 neuronal cells as the large concentrations of both compounds when they use along. This concentration reduction of HBPCD or MBCD needed for the treatment of NPC1 in combination with low concentration of delta-tocopherol may be important for the clinical use in patients.

Dosage recommendations taken from the studies using human NPC1 neutral stem cells (NPSCs) are as follows: (1) The IC50 value for HBPCD on reduction of cholesterol accumulation measured by the Amplex-red cholesterol assay is 50 uM and IC50 for delta-tocopherol is 15 uM. Thus, the ratio is 3.3 fold. (2) In combination therapy experiment, 50 uM HBPCD+10 uM delta-tocopherol significant reduced cholesterol accumulation that is comparable with 5 mM HBPCD, while 50 uM HBPCD or 10 uM delta-tocopherol along did not show the significant cholesterol reduction effect. (3) The molecule weight ratio of HBPCD (MW=1380.25) and delta-tocopherol (MW=402.65) is 3.4 fold.

Given a mouse body weight of 25 g, where brain:body ratio is 1:40, and assuming a complete distribution of HBPCD and delta-tocopherol after direct central envious system injection of compound by intracerebroventricular injection or intrathecal injection, a preferred single use of cyclodextrin for mammals including humans is from 0.1 mg/Kg to 8 mg/Kg, more preferably 0.5 mg/kG or 1.0 mg/Kg to 2 mg/Kg, 3 mg/Kg, 4 mg/Kg, 5 mg/Kg, 6 mg/Kg, 7 mg/Kg or 84 mg/Kg. One specific preferred single use of cyclodextrin for mammals including humans is 3 mg/Kg. In a combination therapy of a cyclodextrin compound administered together or otherwise in conjuction with a vitamin E compound such as delta-tocopherol, a preferred single dose for a mammal including a human may be from 0.05 to 1 mg/kg for each of the cyclodextrin compound and vitamin E compound (such as delta-tocopherol), more preferably 0.1 mg/Kg to 0.5 mg/Kg, 0.6 mg/Kg or 0.7 mg/Kg for each of a cyclodextrin compound and vitam E compound such as delta-tocopherol, still more preferably 0.1 mg/Kg to 0.3 mg/Kg or 0.4 mg/Kg for each of a cyclodextrin compound and vitam E compound such as delta-tocopherol.

Example 6 Effects of Cyclodextrin Single Use and Combination Therapy of Cyclodextrin with δ-Tocopherol

The effects of cyclodextrin in single use and in combination therapy with delta-tocopherol have been determined in patient derived skin fibroblasts with nine types of lysosomal storage diseases including NPC1 (FIG. 14), Batten, Farber, ML III (FIG. 15), MLIV (FIG. 16), MPS1 (FIG. 17), MPS VI (FIG. 18), NPA and Wolman disease. It was found that for single compound use, 8 mM HBPCD or 300 uM MBCD were needed for the significant effect on reduction of the enlarged lysosome size in those cells. However, in a combination with 10 uM delta-tocopherol, 500 uM HBPCD or 20 uM MBCD significantly reduced enlarged lysosomes in these cells. The results indicate an additive/synergistic effect of cyclodextrin with delta-tocopherol on reduction of enlarged lysosomes in the primary fibroblasts derived from patients with those nine lysosomal storage diseases. The results also indicate that the dose of cyclodextrin can be reduced 10 fold or more when it is used in combination with delta-tocopherol. The significant reduction of cyclodextrin dose in combination with delta-tocopherol is important for the treatment of lysosomal storage diseases because the high dose of cyclodextrin may cause server side effects in prolonged treatment process (it is possible that many of these patients may need a life time treatment).

In addition, the high plasma and brain concentrations of cyclodextrin are difficult to achieve in the treatment of LSD patients. The 10 fold reductions of cyclodextrin concentration required in the combination therapy with delta-tocopherol makes the clinical use of cyclodextrin in LSD patients more feasible. Furthermore, delta-tocopherol is difficult to be dissolved in aqueous solution for use in patients that can be resolved in the combination therapy because cyclodextrin can facilitate delta-tocopherol dissolving in solution.

Below is Table 1, showing the cell lines used for the above referenced studies.

TABLE 1 Eponym Coriell of Disease Affected Accumulated Catalog disease name Abbreviation gene Protein Lipid Genotype # Batten Ceroid CLN2 TPP1 Tripeptidyl lipopigments p.R127X, GM16485 lipofuscinosis, peptidase I (lipofuscin) p.R208X neuronal 2 Fabry Alpha- GLA Alpha globotriaosylceramide p.W162X, GM00107 galactosidase galactosidase A rs2071397 A deficiency rs2071228 Farber Lipogranulomatosis AC Acid ceramidase ceramide p.Y36C, GM20015 (N-acylsphingosine p.Y36C amidohydrolase) Nieman NPC1 NPC1 NPC1 Unesterified p.P237S, GM03123 n-Pick, cholesterol p.I1061T type C1 Nieman NPC2 NPC2 NPC2 Unesterified p.C93F, GM17910 n-Pick, cholesterol p.C93F type C2 Nieman NPA ASM Acid Sphingomyelin p.L302P, GM16195 n-Pick, sphingomyelinase p.L302P type A Sanfilippo Mucopolysaccharidosis MPS NAGL N-acetyl-alpha-D- Partially degraded p.R297X, GM02552 type III type B IIIB U glucosaminidase heparan sulfate p.R643H B Tay- GM2 TSD Beta GM2 ganglioside c.1278ins GM00221 Sachs gangliosidosis hexosaminidase TATC A c.1278ins TATC Wolman Lysosomal LAL Lysosomal acid Cholesteryl ester unknown GM11851 acid lipase lipase & triglycerides deficiency

The contents of all references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated herein in their entireties by reference. Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art.

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. Such equivalents are intended with be encompassed by the following claims.

Claims

1. A method of treating a lysosomal storage disorder in a subject, comprising:

determining that the subject is in need of non-cholesterol lipid reduction or reduction of non-cholesterol dominant lipid and macromolecule accumulation, and
administering to the subject in need thereof; an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

2-4. (canceled)

5. The method of claim 1, wherein the non-cholesterol lipid is lipopigments, globotriaosylceramide, ceramide, sphingomyelin, heparan sulfate, partially degraded heparan sulfate, GM2 ganglioside, triglycerides, or cholesterol esters.

6. The method of claim 1, wherein the lysosomal storage disorder is Tay-Sachs disease, Sphingolipidoses, Gaucher disease, Mucolipidosis, Galactosialidosis, Salla disorder, Cystinosis, Danon disease, Fabry disease, Farber disease, Lipofuscinoses, Pompe disease, Gangliodisosis, ISSD, Krabbe disease, leukodystrophy, Hurler disease, Scheie disease, Hunter disease, San Filippo disease, Sandhoff disease, Schinder disease, Batten disorder, or Wolman disease.

7. (canceled)

8. The method of claim 1, wherein the cyclodextrin compound is of formula (I): or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, wherein,

each R is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is optionally substituted; or —C(0)ORB, —OC(0)RB, C(0)RB, or —C(0)NRARB;
each R1 is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, halogen, hydroxy, amino, —CN, —CF3, —N3, —NO2, —ORB, —SRB, —SORB, —SO2RB, —N(RB)S(02)—RB, —(RB)S(O2)NRARB, —NRARB, —C(0)ORB, —0C(0)RB, —C(0)RB, —C(0)NRARB, or —N(RB)C(0)RB; each of which is optionally substituted;
each RA is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is optionally substituted;
each RB is independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl, each of which is optionally substituted; n is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and
each m is independently 0, 1, 2, 3, 4, or 5.

9. The method of claim 8, wherein each R is independently H, optionally substituted alkyl, —C(0)ORB, -0C(0)RB, —C(0)RB, or —C(0)NRARB.

10. The method of claim 9, wherein each R is independently H, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, or octyl; wherein each is straight chain or branched.

11. The method of claim 8, wherein n is 1,2, or 3.

12. The method of claim 1, wherein the cyclodextrin is 2-hydroxypropyl-β-cyclodextrin (2HPβCD), hydroxypropyl-β-cyclodextrin (HPβCD), methyl-β-cyclodextrin (MβCD), α-cyclodextrin, β-cyclodextrin, or γ-cyclodextrin, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof.

13. The method of claim 1, further comprising the step of administering an additional therapeutic agent distinct from the cyclodextrin compound.

14. The method of claim 13 wherein the additional therapeutic agent is administered in combination or otherwise in conjunction with the cyclodextrin compound.

15. The method of claim 13 wherein the additional therapeutic agent is a vitamin.

16. The method of claim 15 wherein the additional therapeutic agent is vitamin E.

17-18. (canceled)

19. The method of claim 1 wherein the step of administering the cyclodextrin comprises administering the compound in a dosage of between about 0.01 μg/kg and 100 mg/kg.

20-35. (canceled)

36. A method of reducing non-cholesterol lipids or reduction of non-cholesterol dominant lipid and macromolecule accumulation in a subject, the method comprising administering to the subject a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof; and detecting the amount of lipid reduction.

37. The method of claim 36 wherein the subject is identified as being in need of lipid reduction.

38. The method of claim 1 further comprising the step of synthesizing or obtaining the cyclodextrin compound.

39. The method of claim 1, wherein the subject is a human.

40. The method of claim 1 wherein the cyclodextrin compound and/or any additional distinct therapeutic agents are administered intracranially to the subject.

41. A pharmaceutical composition comprising a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and vitamin E, together with a pharmaceutically-acceptable carrier or excipient.

42-44. (canceled)

45. A kit comprising an effective amount of a cyclodextrin compound, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in unit dosage form, together with instructions for administering the compound to a subject suffering from a lysosomal storage disorder.

46-48. (canceled)

Patent History
Publication number: 20150216895
Type: Application
Filed: Aug 3, 2013
Publication Date: Aug 6, 2015
Inventors: John McKew (Boyds, MD), Wei Zheng (Potomac, MD), Miao Xu (Hangzhou), Manju Swaroop (Gaithersburg, MD), Juan J. Marugan (Gaithersburg, MD)
Application Number: 14/419,471
Classifications
International Classification: A61K 31/724 (20060101); C08B 37/16 (20060101); G01N 33/92 (20060101); A61K 31/355 (20060101);