Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ORAL FORMULATIONS OF BIPOLAR TRANS CAROTENOIDS
Document Type and Number:
WIPO Patent Application WO/2011/152869
Kind Code:
A1
Abstract:
The subject invention relates to a variety of formulations of bipolar trans.carotenoids including pharmaceutical compositions for oral delivery of a bipolar trans carotenoid comprising i) a bipolar trans carotenoid, ii) a cyclodextrin, and iii) a coating. The invention also relates to preparation of such formulations and their uses.

Inventors:
GAINER JOHN L (US)
MURRAY ROBERT (US)
Application Number:
PCT/US2011/000997
Publication Date:
December 08, 2011
Filing Date:
June 02, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DIFFUSION PHARMACEUTICALS LLC (US)
GAINER JOHN L (US)
MURRAY ROBERT (US)
International Classes:
A01N37/00; A23D9/00; A01N43/04; A61K31/715
Foreign References:
US20060276372A12006-12-07
US20040116729A12004-06-17
US20090110746A12009-04-30
US20040109920A12004-06-10
US20080113031A12008-05-15
US20090176287A12009-07-09
US20090169586A12009-07-02
US7446101B12008-11-04
Other References:
See also references of EP 2575487A4
Attorney, Agent or Firm:
BYERS, Duane, M. (901 North Glebe Road 11th Floo, Arlington VA, US)
Download PDF:
Claims:
What is claimed is:

1 . A pharmaceutical composition for oral delivery of a bipolar trans carotenoid comprising: i) a bipolar trans carotenoid,

ii) a cyclodextrin, and

iii) a coating.

2. A pharmaceutical composition as in claim 1 wherein the bipolar trans carotenoid is a salt.

3. A pharmaceutical composition as in claim 1 wherein the bipolar trans carotenoid is TSC.

4. A pharmaceutical composition as in claim 1 wherein the coating is an enteric coating.

5. A pharmaceutical composition as in claim 1 wherein the coating is an enteric coating which will release the bipolar trans carotenoid at pH greater than 5.5.

6. A pharmaceutical composition as in claim 1 wherein the coating is an enteric coating which will release the bipolar trans carotenoid at pH greater than 6.5.

7. A pharmaceutical composition as in claim 1 wherein the coating is a coating which will release the bipolar trans carotenoid in the intestines.

8. A pharmaceutical composition as in claim 1 wherein the coating is an acrylate polymer.

9. A pharmaceutical composition as in claim 1 wherein the cyclodextrin is selected from the group consisting of alpha cyclodextrin, beta cyclodextrin, and gamma cyclodextrin.

10. A pharmaceutical composition as in claim 1 wherein the bipolar trans carotenoid is synthetic trans sodium crocetinate wherein the purity level of the trans isomer in the composition is such that under UV-visible analysis, the absorbency of the highest peak which occurs in the visible wave length range divided by the absorbency of the peak which occurs in the UV wave length range is greater than 7.5.

1 1 . A pharmaceutical composition as in claim 1 wherein said composition is in unit dosage form.

12. A pharmaceutical composition as in claim 1 wherein the composition is in the form of a tablet, pill, or capsule.

13. A method of forming a pharmaceutical composition for oral delivery of a bipolar trans carotenoid comprising:

mixing a bipolar trans carotenoid with a cyclodextrin,

adding the mixture to a capsule or making a tablet, and

adding an enteric coating.

14. A method of increasing the diffusivity of oxygen in a mammal comprising administering orally to a mammal a therapeutically effective amount of the composition of claim 1 .

15. A method of treating a mammal having a disease or condition characterized by hypoxia comprising administering orally to such mammal a therapeutically effective amount of the composition of claim 1 .

16. A method of treating a mammal at risk of a disease or condition characterized by hypoxia comprising administering to such mammal a therapeutically effective amount of the composition of claim 1.

1 7. A method as in claim 1 5 or 16 wherein said disease or condition characterized by hypoxia is selected from the group consisting of ischemia, cancer, traumatic brain injury, respiratory disease, hemorrhagic shock, cardiovascular disease, multiple organ failure, atherosclerosis, PAD, PVD, myocardial infarction, emphysema, asthma, ALI, ARDS, COPD, hypertension, cerebral edema, papillomas, spinal cord injury, stroke, and conditions of the central nervous system.

18. A method of treating a mammal having metabolic syndrome comprising administering to such mammal a therapeutically effective amount of the composition comprising a bipolar trans carotenoid.

19. A method of treating a mammal having cancer comprising administering to such mammal a therapeutically effective amount of the composition consisting essentially of a bipolar trans carotenoid salt.

20. A method of treating a mammal having a neurodegenerative disease selected from the group consisting of Parkinson's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, Huntington's disease, Lewy body disease, and spinal muscular atrophy, comprising

administering to such mammal a therapeutically effective amount of the composition comprising a bipolar trans carotenoid.

21 . A method as in claim 1 8 or 20 wherein the bipolar trans carotenoid is a bipolar trans carotenoid salt.

22. A method as in claim 19 or 21 wherein the composition further comprises a cyclodextrin.

23. A method as in claim 1 8, 19 or 20 wherein the composition is administered orally.

Description:
Oral Formulations of Bipolar Trans Carotenoids

This application claims priority from U.S. provisional patent application number 61/350,804, filed on June 2, 2010, the entire contents of which are hereby incorporated by reference.

The subject invention relates to formulations of diffusion enhancing compounds. The compositions of the subject invention typically include a bipolar trans carotenoid, a

cyclodextrin and a coating. Included are compositions that are enterically coated with a pH responsive compound for oral delivery.

Background of the Invention

Peroral delivery of therapeutics is generally considered to be the most popular method of drug delivery for patients since this route, in general, increases patient compliance, decreases the number of side effects associated with injections, and provides convenience for the user. Such an administration route is greatly favored for dosing chronically ill patients.

* * *

Carotenoids are a class of hydrocarbons consisting of isoprenoid units. The backbone of the molecule consists of conjugated carbon-carbon double and single bonds, and can also have pendant groups. Crocetin and trans sodium crocetinate (TSC) are known to increase the diffusivity of oxygen in aqueous solutions.

US Patent 6,060,5 1 1 relates to trans sodium crocetinate (TSC) and its uses. The patent covers various uses of TSC such as improving oxygen diffusivity and treatment of hemorrhagic shock.

US Patent application Ser. No. 10/647, 132 relates to synthesis methods for making bipolar trans carotenoid salts (BTC) and methods of using them. US Patent application Ser. No. 1 1 /361 ,054 relates to improved BTC synthesis methods and novel uses of the BTC.

US Patent application Ser. No. 12/081 ,236 relates to the use of bipolar trans carotenoids as a pretreatment and in the treatment of peripheral vascular disease.

US Patent Application Ser. No. 12/289,713 relates to a new class of therapeutics that enhance small molecule diffusion.

US Provisional Application Ser. No. 61/213,575 relates to the use of diffusion enhancing compounds with thrombolytics.

A variety of bipolar trans carotenoids formulations have been disclosed. See commonly owned application US Patent application Ser. No. 10/647, 132 and US Patent application Ser. No. 1 1/361 ,054.

Summary of the Invention

The pharmaceutical compositions of the subject invention include a diffusion enhancing compound, a cyclodextrin and a coating. The invention also relates to methods of forming a pharmaceutical composition for oral delivery of a bipolar trans carotenoid comprising mixing a bipolar trans carotenoid with a cyclodextrin, adding the mixture to a capsule or making a tablet, and adding coating, advantageously, an enteric coating. Also included in the invention are methods of increasing the diffusivity of oxygen in a mammal and methods of treating a mammal having a disease or condition characterized by hypoxia such as ischemia, cancer, traumatic brain injury, respiratory disease, hemorrhagic shock,

cardiovascular disease, multiple organ failure, atherosclerosis, PAD, PVD, myocardial infarction, emphysema, asthma, AL1, ARDS, COPD, hypertension, cerebral edema, papillomas, spinal cord injury, conditions of the central nervous system particularly diseases characterized by neuro-degeneration, and metabolic syndrome and its.complications.

Detailed Description of the Invention

Although a bipolar trans carotenoid (BTC) such as trans sodium crocetinate (TSC), is a member of the carotenoid family of compounds, it cannot be orally dosed in a manner similar to other carotenoids which are sold in capsule or pill form (e.g., beta carotene or Vitamin A). Two factors which must be accounted for in developing an oral formulation of a BTC compound are pH and solubility considerations which are totally different from other carotenoid compounds. Methods used for formulation of other carotenoid compounds simply don't work for compounds such as TSC.

TSC is precipitated under acidic conditions, such as those which exist in the stomach. This solid material is practically insoluble in the acidic environment of the stomach. TSC is not stable in acidic conditions and will be converted to the cis isomer which can precipitate. A system has been devised that will protect TSC in the harsh, acidic environment of the stomach and release TSC in a more favorable pH region. A more favorable pH does exist in the small intestines. In-sit closed loop studies have shown increased bioavailability when TSC is administered directly to the small intestine compared to the stomach. See Examples below. TSC's effectiveness is believed to be dependant on the TSC concentrations levels found in the body. The effectiveness of TSC can be prolonged by administering TSC orally.

Tablets of bipolar trans carotenoids include excipients that are commonly used in making tablets. Advantageously, tablets also have an enteric coating placed on them, so that they will dissolve in the higher pH (> 5.5) areas of the digestive track instead of in the acidic stomach.

Compositions of the Invention The compositions of the subject invention are all manufactured to be pharmaceutical grade, i.e. pharmaceutical compositions. Such formulations can include pharmaceutically acceptable carriers known to those skilled in the art as well as other therapeutic agents.

Advantageously, the formulation does not include a compound that inhibits the ability of the diffusing enhancing compound to improve diffusivity.

The compositions of the subject invention include a) a diffusion enhancing compound, b) a cyclodextrin and c) a coating. As an alternative, a pH responsive carrier (i.e. TSC is dispersed/held in a polymer matrix), or a time release system (also known as sustained release, controlled, etc.) can be used.

A. Diffusion Enhancing Compounds

The diffusion enhancing compounds of the invention include those compounds described in US Ser. No. 10/647, 132, US Ser. No. 1 1 /361 ,054, US Ser. No. 12/081 ,236 and US Ser. No. 12/289,713, each of which is hereby incorporated by reference in its entirety.

The diffusion enhancing compounds of the subject invention are trans carotenoids including trans carotenoid diesters, dialcohols, diketones and diacids, bipolar trans carotenoids (BTC), and bipolar trans carotenoid salts (BTCS). Included are bipolar trans carotenoid compounds having the formula:

YZ-TCRO-ZY

where:

Y = a cation

Z = a polar group which is associated with the cation, and

TCRO = trans carotenoid skeleton, such as TSC.

More specifically:

Y (which can be the same or different at the two ends) =H or a cation other than H, preferably Na + or K + or Li + . Y is advantageously a monovalent metal ion. Y can also be an organic cation, e. g., R 4 N + , R 3 S + , where R is H, or C n H2 n +i where n is 1 -10,

advantageously 1 -6. For example, R can be methyl, ethyl, propyl or butyl.

Z (which can be the same or different at the two ends)= polar group which is associated with H or the cation. Optionally including the terminal carbon on the carotenoid (or carotenoid related compound), this group can be a carboxyl (COO ) group or a CO group (e.g. ester, aldehyde or ketone group), or a hydroxyl group. This group can also be a sulfate group (OS0 3 " ) or a monophosphate group (OP0 3 ~ ),

(OP(OH)0 2 " ), a diphosphate group, triphosphate or combinations thereof. This group can also be an ester group of COOR where the R is

TCRO = trans carotenoid or carotenoid related skeleton

(advantageously less than 100 carbons) which is linear, has pendant groups (defined below), and typically comprises "conjugated" or alternating carbon-carbon double and single bonds (in one embodiment, the TCRO is not fully conjugated as in a lycopene). The pendant groups (X) are typically methyl groups but can be other groups as discussed below. In an advantageous embodiment, the units of the skeleton are joined in such a manner that their arrangement is reversed at the center of the molecule. The 4 single bonds that surround a carbon-carbon double bond all lie in the same plane. If the pendant groups are on the same side of the carbon-carbon double bond, the groups are designated as cis (also known as "Z"); if they are on the opposite side of the carbon- carbon bond, they are designated as trans (also known as "E").

Throughout this case, the isomers will be referred to as cis and trans.

The compounds of the subject invention are trans. The cis isomer typically is a detriment— and results in the diffusivity not being increased. In one embodiment, a cis isomer can be utilized where the skeleton remains linear. The placement of the pendant groups can be symmetric relative to the central point of the molecule or can be asymmetric so that the left side of the molecule does not look the same as the right side of the molecule either in terms of the type of pendant group or their spatial relationship with respect to the center carbon.

The pendant groups X (which can be the same or different) are hydrogen (H) atoms, or a linear or branched hydrocarbon group having 10 or less carbons, advantageously 4 or less, (optionally containing a halogen), or a halogen. X could also be an ester group (COO-) or an ethoxy/methoxy group. Examples of X are a methyl group (CH3), an ethyl group (C2H5), a phenyl or single aromatic ring structure with or without pendant groups from the ring, a halogen-containing alkyl group (C 1 -C 10) such as CH 2 C1, or a halogen such as CI or Br or a methoxy (OCH3) or ethoxy (OCH 2 CH 3 ). The pendant groups can be the same or different but the pendant groups utilized must maintain the skeleton as linear.

Although many carotenoids exist in nature, carotenoid salts do not. Commonly-owned US Patent 6,060,51 1 hereby incorporated by reference in its entirety, relates to trans sodium crocetinate (TSC). The TSC was made by reacting naturally occurring saffron with sodium hydroxide followed by extractions that selected primarily for the trans isomer. The presence of the cis and trans isomers of a carotenoid or carotenoid salt can be determined by looking at the ultraviolet-visible spectrum for the carotenoid sample dissolved in an aqueous solution. Given the spectrum, the value of the absorbance of the highest peak occurs in the visible wave length range of 380 to 470 nm (the number depending on the solvent used and the chain length of the BTC or BTCS). The addition of pendant groups or differing chain lengths may change this peak absorbance but someone skilled in the art will recognize the existence of an absorbance peak in the visible range corresponding to the conjugated backbone structure of these molecules, divided by the absorbency of the peak which occurs in the UV wave length range of 220 to 300 nm, can be used to determine the purity level of the trans isomer. When the trans carotenoid diester (TCD) or BTCS is dissolved in water, the highest visible wave length range peak will be at between 380 nm to 470 nm (depending on the exact chemical structure, backbone length and pendant groups) and the UV wave length range peak will be between 220 to 300 nm. According to M. Craw and C. Lambert, Photochemistry and Photobiology, Vol. 38 (2), 241 -243 ( 1983) hereby incorporated by reference in its entirety, the result of the calculation (in that case crocetin was analyzed) was 3.1 , which increased to 6.6 after purification.

Performing the Craw and Lambert analysis on the synthetic TSC as described in US Ser. No. 10/647, 1 32 and US Ser. No. 1 1 /361 ,054, that ratio is greater than 7.0 (e.g. 7.0 to 8.5), advantageously greater than 7.5 (e.g. 7.5 - 8.5), most advantageously greater than 8. The synthesized material is a "purer" or highly purified trans isomer.

Advantageously, the trans carotenoid is crocetin, crocin, a bipolar trans carotenoid (BTC) salt such as TSC, or a carotenoid diester, alcohol or acid.

B. Cyclodextrins Many excipients have been suggested to increase bioavailability of drugs from the gastrointestinal tract such as surfactants, chelating agents, glycols, polyethylene glycol and others; however, cyclodextrin with a bipolar trans carotenoid works extremely well.

A detailed description of cyclodextrins in combination with bipolar trans carotenoids can be found in commonly owned application US Ser. No. 1 1 /361 ,054 which is hereby incorporated by reference in its entirety.

Advantageously, the bipolar trans carotenoid is in the form of a composition

comprising a trans carotenoid and a cyclodextrin, for example alpha cyclodextrin, beta cyclodextrin or gamma cyclodextrin. The cyclodextrin can be hydroxylpropyl-beta- cyclodextrin or 2-hydroxylpropyl-gamma-cyclodextrin. In another embodiment, the

composition further comprises mannitol or saline. In a still further embodiment, the

composition further comprises a compound to regulate pH such as bicarbonate or glycine.

Advantageously, the ratio of a bipolar trans carotenoid to the cyclodextrin is up to 1 : 10. Advantageously, up to 1 :4, e.g. 2: 1 , 1 : 1 , or 1 :4.

C. Coatings

An enteric coating is a barrier applied to oral medication that controls the location in the digestive system where it is absorbed. Enteric refers to the small intestine, therefore enteric coatings prevent release of medication before it reaches the small intestine. Most enteric coatings work by presenting a surface that is stable at the highly acidic pHfound in the stomach, but breaks down rapidly at a less acidic (relatively more basic) pH. For example, they will not dissolve in the acidic juices of the stomach (pH of 1 to 3), but they will in the higher pH(above pH 5.5) environment present in the small intestine. Materials used for enteric coatings include fatty acids, waxes, and shellac as well as plastics.

Bipolar trans carotenoids precipitate in acid conditions. For such types of drugs, an enteric coating can be added to the formulation to protect the active substance from the stomach's acidic exposure, delivering the active instead to a basic pH environment (intestine's pH 5.5 and above) where it is more soluble, and can give its desired action. TSC's effectiveness is believed to be dependent on the TSC concentrations levels found in the body. The effectiveness of TSC can be prolonged by administering TSC orally. Since TSC precipitates in acidic conditions, the compositions of the subject invention protect TSC in the harsh, acidic environment of the stomach and release TSC in a more favorable pH region in the intestines.

An enteric coating is applied to oral dosage formulations in order to protect the active substance from dissolution in the gastric fluid within the stomach. The most common reasons for using an enteric coating include:

• Protection of the active substance from the gastric enzymes or acidity of the gastric fluid

• Masking of the task or odor

• Preventing irritation of the stomach including nausea and vomiting

• Sustained release for controlled absorption

• Delivery of the active substance to a specific site in the digestive tract that is more favorable for systemic absorption

Another embodiment includes using an enteric coating that responds at a broader range of pH values in order to allow a bipolar trans carotenoid such as TSC to absorb in more sections of the intestines, thus increasing the surface area available for absorption.

Composition of Coatings

There are many materials available that are used to enterically coat materials. Most function by either a slow erosion of the coating material (carnauba wax, keratin, gluten, etc.) or by a pH responsive coating. Materials relying on the erosion mechanism are dependent on gastric emptying times. The second type, the pH responsive coating, are hydrophobic and water insoluble at low pH conditions and become soluble at higher pHs. Thus, the coating is insoluble at the harsh acidic environment of the stomach and dissolves at the higher pH region of the intestine. There are many pH sensitive polymers available which can be selected based on a specific pH at which disintegration should occur. The table below lists some of the most common pH sensitive polymers.

Common pH sensitive polymers used for enteric coatings

Cellulose acetate phthalate (CAP)

Cellulose acetate succinate

Acrylate polymers

Hydroxy propyl methyl cellulose phthalate

Hydroxy propyl methyl cellulose acetate succinate

Polyvinyl acetate phthalate

Three commercially available acrylate polymers—copolymers derived from esters of acrylic and methacrylic acid are Eudragit L30D-55, L I 00, and FS30D. The pH-dependent functionality of these polymers are determined by their functional, carboxylic acid groups:

Eudragit L30D-55 : Dissolution occurs at a pH of 5.5 or greater with a targeted release area of the duodenum.

Eudragit L I 00: Dissolution occurs at a pH above 6.0 with a targeted release area of the

Eudragit FS30D: Dissolution occurs at a pH above 7.0 with a targeted release area of the colon.

D. Other Ingredients

Agents can be added to buffer the diffusion enhancing compounds. Other agents can be added to affect the osmolality, or added as compounding agents needed for oral formulations. Specific buffering agents include glycine, sodium carbonate, sodium bicarbonate, magnesium carbonate, and aluminum hydroxide. Specific agents to affect osmolality include mannitol, and polysaccharides.

Other agents that can be added include: Prosolv 90 is a silicified microcrystalline cellulose. The PROSOLV SMCC 90 is said to offer a balance of best in class compaction and flow for tablet formulations. It improves formulation flow, enhances consolidation of the particles, and improves content uniformity.

Crospovidone XL 90 is a cross-linked polyvinylpyrrolidone which is used in tableting as a spheronization aid and an alternative to microcrystalline cellulose. Spheronization, marmumerization, pelletization and micropelletization all mean the same thing: the manufacture of products in small spheres for use in pharmaceutical and related industries. All the products produced by these processes can be called spheroids, spheres, micro-spheres, pellets, micro-pellets or pharmaceutical pellets. All these terms refer to the same thing. Size range is normally from about 0.8 mm to 1.5 mm in diameter although smaller and larger are possible. In contrast, the term granulation normally refers to irregularly shaped particles with a large size range within a batch. Granulated materials are generally less dense.

Magnesium stearate, also called octadecanoic acid, magnesium salt, is a white substance which is solid at room temperature. It has the chemical formula C 36 H 7 oMg0 4 . It is a salt containing two equivalents of stearate (the anion of stearic acid) and one magnesium cation (Mg 2+ ).

Magnesium stearate melts at about 88 °C, is not soluble in water, and is generally considered safe for human consumption. Because it is widely regarded as harmless, it is often used as a filling agent in the manufacture of medical tablets and capsules. In this regard, the substance is also useful because it has lubricating properties, preventing ingredients from sticking to manufacturing equipment during the compression of chemical powders into solid tablets.

Kollidon is a polyvinyl polymer of variable molecular weight; used as a suspending and dispersing agent and vehicle for pharmaceuticals.

Methods of Formulation

Formulations of the present invention suitable for oral administration can be presented as discrete units such as pills, capsules, cachets or tablets, as powder or granules, or as a solution, suspension or emulsion. Formulations suitable for oral administration further include lozenges, and pastilles. The formulations can conveniently be presented in unit dosage form, and can be prepared by methods known in the art of pharmacy. The formulation can be for immediate, or slow or controlled release of the diffusion enhancing compound. The advantages of a sustained release system (also known as time release, controlled release, etc.) are that dosing frequency can decrease and the systemic drug concentrations are steadier for a longer duration as compared to other formulations of the same drug.

Appropriate dosages of the compositions of the invention will depend on the metabolism of the given compound, and the severity of the condition being treated. For a dose to be "therapeutically effective", it must have the desired effect, i.e. it must relieve symptoms of the indication for which it is given. The therapeutically effective dosage will depend upon the condition treated, the severity of the condition, the stage and individual characteristics of each mammalian patient addressed, and the clearance of the diffusion enhancing effect.

Typically the compositions of the invention are made by mixing the bipolar trans carotenoid and the selected cyclodextrin at a ratio of up to 1 : 10. Mixing is done by any pharmaceutically accepted method. The mixture is then either loaded into a capsule container or stamped into a tablet (which also can contain the ingredients mentioned previously to promote release from the molds, etc.). The capsules or tablets are then coated by pH-sensitive polymer such as a Eudragit in such a manner so as to create a continuous coating.

In one embodiment of the invention, multiple types of enterically coated beads are placed in a capsule or other system for oral delivery. The beads are composed of a first portion or group of beads having a first coating and a second portion or group of beads having a second coating. Additional groups of beads with different coatings can also be added. An example is a capsule containing beads having three different types of Eudragit coatings that release at different pH values and release over a longer period of time than a capsule containing only one type of bead. Therapeutic Uses and Modes of Administration

The compositions of the invention have therapeutic uses in treating mammals having tissues experiencing low oxygen levels (hypoxia) or in various conditions involving the central nervous system.

The uses of the compositions of the invention include those disclosed in commonly owned US patent 6,060,51 1 , US Patent application Ser. No. 10/647, 132, US Patent application Ser. No. 1 1 /361 ,054, US Patent application Ser. No. 12/081 ,236 and US provisional Patent application Ser. No. 61 /213,575, each of which is hereby incorporated by reference in its entirety.

The oral compositions of the invention are useful in the treatment of:

hemorrhagic shock, respiratory disease, asthma, emphysema, ALI, ARDS, COPD ischemia, cardiovascular disease, atherosclerosis, myocardial infarction, hypertension, ventricular fibrillation stroke, traumatic brain injury, cerebral edema,

conditions of the central nervous system (Alzheimer's disease, Parkinson's disease, and other neurodegenerative diseases) Neurodegeneration is the umbrella term for the progressive loss of structure or function of neurons, including death of neurons.

Examples of degenerative nerve diseases include: Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, Huntington's disease, Lewy body disease, and spinal muscular atrophy. See Example 7. arthritis,

anemia, (anemia of prematurity, Fanconi anemia, hymolytic anemia, microcytic anemia, a normochromic anemia, a macrocytic anemia, hereditary spherocytosis, sickle-cell anemia, warm autoimmune hemolytic anemia, cold agglutinin hemolytic anemia), chronic renal failure, hypertension, papillomas, spinal cord injuries, cancer (advantageously as an adjunct to i) radiation therapy including external beam radiation, gamma knife, brachytherapy, tomotherapy, and proton beam, including fractionated, 3D conformal radiotherapy, intracavitary radiation, and intensity modulated radiotherapy (IMRT), and/or ii) chemotherapy including temozolimide). diabetes, diabetic retinopathy, peripheral vascular disease/claudication , embolism, blood clot, spinal

stenosis/neurogenic claudication, diseases where organs do not get enough oxygen such as Wegener's granulomatosis

The compositions of the invention are also useful as a pretreatment or for treating mammals at risk for the above-noted diseases/conditions.

The compositions are also useful in neuroprotection, i.e. in preventing or delaying the complications associated with neurodegenerative disorders such as Parkinsons disease or Alzheimers disease. They are also useful in reducing the amount of ischemia resulting from surgery in a mammal by administering the composition before during or after surgery The compositions are also useful in enhancing performance when respiration/exertion is increased or stressed, in increasing aerobic metabolism, and in increasing endurance during physical activity such as running walking or lifting.

For the following uses, the diffusion enhancing compounds are administered by any suitable route including oral, nasal or inhalation, topical, parenteral (including subcutaneous, intramuscular, intravenous, intradermal, transdermal and intraosseus), vaginal or rectal. The preferred route of administration will depend on the circumstances. An inhalation route, or intravenous or intramuscular injection is advantageous for treatment in emergency situations, where it is necessary for the diffusion enhancing compound to enter the bloodstream very quickly. In one embodiment, a composition of a cyclodextrin and bipolar trans carotenoid dissolved in sterile water can be injected, either intramuscularly (IM) or intravenously (IV). The formulations thus include those suitable for administration through such routes (liquid or powder to be nebulized). It will be appreciated that the preferred route may vary, for example, with the condition and age of the patient. critical limb ischemia

Parkinson's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, Huntington's disease, Lewy body disease, and spinal muscular atrophy, multiple sclerosis, metabolic syndrome peripheral neuropathy cerebral palsy cancer - a bipolar trans carotenoid salt, such as TSC, is the chemotherapy used to cause regression of many types of cancerous tumors, i.e. without use of radiation or other chemotherapy. Example 8 relates to treating cancerous tumors with TSC. TSC does not work by killing the cancer cells, but, while not wishing to be bound by theory, is thought to work by causing the cells to revert to more mature (and thus, more nearly normal) cells. The use of retinoids (such as all trans retinoic acid), or their salts, for the treatment of cancer is excluded from the invention. Examples of the types of cancer/tumors which can be treated are: skin, lung, breast, brain, bladder, prostate and colon cancers/tumors.

In one embodiment, more than one diffusion enhancing compound is administered. Alternatively, hemoglobins or fluorocarbons and a diffusion enhancing compound can be given together.

* * *

The following Examples are illustrative, but not limiting of the compositions and methods of the present invention. Other suitable modifications and adaptations of a variety of conditions and parameters normally encountered which are obvious to those skilled in the art are within the spirit and scope of this invention.

EXAMPLES

Example 1

Tablet Composition (without cyclodextrin)

Two formulations were made that consist of the following ingredients:

65% Blend

65.0% TSC

26.0% Prosolv 90

2.9% Crospovidone XL 90

1.5% Magnesium Stearate 4.6% Kollidon

60% Blend

60.4% TSC

30. 1 % Prosolv 90

3.8% Crospovidone XL 90

1 .4% Magnesium Stearate

4.3% Kollidon

The TSC tablets were made in three (3) steps:

Step 1 : A mixture containing 70% TSC, 25% Prosolv 90 and 5% Kollidon 25 were mixed in a bag for 5 minutes and roll compacted using a Vector roller compactor. The conditions for the roller compactor were roll pressure = 800 psi, screw speed = 10 rpm, roll speed = 0.95 rpm. This produced a blend called TSC Blend 1.

Step 2: The TSC Blend I from Step 1 was roll compacted using Crospovidone XL 10 and magnesium stearate. The percentages were 97.5% TSC Blend I, 2% Crospovidone XL 10 and 0.5% magnesium stearate. The product of this step is called TSC Granules II.

Step 3: The ribbons from Step 2 were hand crushed and sieved through 20 mesh screen. The granules obtained were mixed with excipients (see below) to get final blends containing 65% TSC and 60% TSC.

65% TSC Blend 60% TSC Blend

95.2% TSC Granules II 88.5 % TSC Granules II

2.8 % Prosolv 90 8.5 % Prosolv 90

1 % Crospovidone XL 2% Crospovidone XL

1 % Magnesium Stearate 1 % Magnesium Stearate Step 4: The tablets were coated with CAP (cellulose acetate phthalate)

To demonstrate the difference in absorption in the stomach and in the intestine, the following studies were performed.

* * *

Example 2

Absorption of TSC Solutions

Male Sprague-Dawley (SD) and Wistar rats weighing approximately 300-400g each were fasted for 24 hours prior to each experiment. Water was given ad libitum and coprophagy was prevented by using cages with wire-mesh floors. Anesthesia was induced and maintained with isoflurane. The carotid artery was exposed and cannulated with PE-50 tubing. The cannula was secured using silk sutures and 2% lidocaine was applied to the wound.

Following cannulation, animals were placed into one of two groups: administration of a TSC dosage formulation directly into the 1 ) stomach via gavage; or 2) into an isolated intestinal segment. For dosing into an isolated intestinal segment, a front midline in was made, exposing the abdomen and intestinal segments. In this study, the ileum was isolated and cannulated with PE-50 tubing, then washed with normal 37°C saline until the washings run clear. The segment was replaced, the abdomen clamped, and the rat was allowed to stabilize for 1 hour. The TSC formulation was then introduced into the isolated segment. The intestine was replaced and the abdomen closed with sutures. Approximately 0.3 mL blood samples were taken for the carotid artery at discrete time intervals following TSC administration. A small amount of sodium heparin was used as an anticoagulant. The blood samples were centrifuged and the resulting plasma volume mixed with 3 volumes of methanol and vortexed. The plasma mixture was then centrifuged and the supernatant was analyzed by HPLC to determine TSC levels. The TSC dosage formulation administered in this study consisted of an intravenous formulation (20 mg/mL TSC, 8% gamma cyclodextrin, 50mM Glycine, and 2.3% Mannitol). Figure 1 on the following page shows that there is an increased bioavailability when TSC is administered directly to the small intestine as compared to the stomach.

time, minutes

Figure 1 . Mean TSC plasma concentration following administration of 2.9 mg/kg TSC directly to the(*) ileum {in-situ dosing) and (O)stomach.

Example 3

Enteric Coated Tablets (without cyclodextrin)

TSC is precipitated and is practically insoluble under the harsh acidic envirnment of the stomach. TSC must be protected from the harsh environment of the stomach and be release in a more favorable, higher pH region of the small intestine. For this study, tablets of TSC were made (Table 1 ) and were enterically coated with either Eudragit L I 00 or cellulose acetate phthalate (CAP). The integrity of both types of protective coatings were confirmed in a USP dissolution study in which enterically coated tablets were first place in a dissolution cell containing simulated gastric fluid (SGF), then transferred to another cell containing simulated intestinal fluid (S1F). The coating and tablet remained intact in the SGF, but dissolution did occur in SIF. This study was performed according to USP protocols.

Table 1. TSC tablet formulation

65% TSC Blend 60% TSC Blend

95.2% TSC Granules II 88.5 % TSC Granules II 2.8 % Prosolv 90 8.5 % Prosolv 90

1 % Crospovidone XL 2% Crospovidone XL

1 % Magnesium Stearate 1 % Magnesium Stearate

These tablets were made using tabletting technology known to one skilled in the art.

Then, in order to examine what happens with enterically-coated TSC, a tablet coated with CAP, and containing 300mg TSC using the 65% blend, was administered orally to dogs. Very low plasma TSC concentrations were found, suggesting that the bioavailability would be too low for clinical use in this formulation.

Example 4

Intestinal Absorption of TSC and Effect of Cvclodextrin

Additional studies were conducted in order to determine the effect on systemic absorption with the addition of gamma cyclodextrin to the TSC.

Male Sprague-Dawley (SD) and Wistar rats weighing approximately 300-400g each were fasted for 24 hours prior to each experiment. Water was given ad libitum and coprophagy was prevented by using cages with wire-mesh floors. Anesthesia was induced and maintained with isoflurane. The carotid artery was exposed and cannulated with PE-50 tubing. The cannula was secured using silk sutures and 2% lidocaine was applied to the wound. Following cannulation, a front midline in was made, exposing the abdomen and intestinal segments. In this study, the jejunum was isolated and the TSC formulation was administered in the proximal jejunum. Approximately 60mg/kg TSC was administered at various gamma cyclodextrin ratios of 0.5 : 1 up to 4: 1 (wt. TSC:wt. gamma cyclodextrin). Movement of TSC within the intestinal segments was not restricted distal to the site of administration. The intestine was replaced and the abdomen closed with sutures.

Approximately 0.3 mL blood samples were taken for the carotid artery at discrete time intervals following TSC administration. A small amount of sodium heparin was used as an anticoagulant. The blood samples were centrifuged and the resulting plasma volume mixed with 3 volumes of methanol and vortexed. The plasma mixture was then centrifuged and the supernatant was analyzed by HPLC to determine TSC levels.

It was found that the γ-cyclodextrin greatly enhances the absorption of the TSC as shown in Figure 3.

Effect of Gamma Cyclodextrin on Absoroption of

TSC into Blood

1 2 3

Weight of CD per weight of TSC

Figure 3: Effect of g-cyclodextrin on TSC absorption in the jejunum of the rat.

Below are the pk curves following in-situ TSC administration into the small intestine. The curves show that cyclodextrin significantly increases intestinal absorption of TSC.

The graph below is TSC plasma concentration ^g/mL) vs. time following administration (min). 90

0 IS 30 45 60 90 120 180 240 300

Example 5

Scalability for Cmax

It has been found that gamma cyclodextrin greatly enhances the systemic absorption of TSC in the Gl tract. The dosage of TSC used in the above study is large, therefore, additional doses of TSC were administered with gamma cyclodextrin at a ratio of 1 : 1 (wt. TSC:wt.

gamma cyclodextrin) in order to determine the scalability of systemic absorption (in terms of Cmax) with respect to TSC dosing amount. For the following study, TSC was administered to the jejunum intestinal segment at the following dosages: 2.5, 5, 10, and 60 mg/kg.

For this study, male Sprague-Dawley (SD) and Wistar rats weighing approximately 300-400g each were fasted for 24 hours prior to each experiment. Water was given ad libitum and coprophagy was prevented by using cages with wire-mesh floors. Anesthesia was induced and maintained with isoflurane. The carotid artery was exposed and cannulated with PE-50 tubing. The cannula was secured using silk sutures and 2% lidocaine was applied to the wound.

Following cannulation, a front midline in was made, exposing the abdomen and intestinal segments. In this study, the jejunum was isolated and the TSC formulation was administered in the proximal jejunum. Movement of TSC within the intestinal segments was not restricted distal to the site of administration. The intestine was replaced and the abdomen closed with sutures. Approximately 0.3 mL blood samples were taken for the carotid artery at discrete time intervals following TSC administration. A small amount of sodium heparin was used as an anticoagulant. The blood samples were centrifuged and the resulting plasma volume mixed with 3 volumes of methanol and vortexed. The plasma mixture was then centrifuged and the supernatant was analyzed by HPLC to determine TSC levels.

The results are shown in Figure 4. The line in Figure 4 is a linear regression of the data, and shows that an excellent fit is obtained. This suggests that the data obtained for the inclusion of γ-cyclodextrin is scalable to other TSC dosages.

Effect of TSC dosage on cyclodextrin

enhancement

40 80

TSC dosage, mg/kg

Figure 4: Effect of TSC dosage in a TSC-y-cyclodextrin mixture on Cmax. Example 6

Intestinal Absorption with and without Enteric Coatings

Additional studies were conducted in order to investigate the benefit of including gamma cyclodextrin with TSC for use in peroral delivery, For this study, TSC and gamma cyclodextrin at a ratio of 1 :4 (wt TSC:wt. gamma cyclodextrin) were packed in size 9 gelatin capsules and either: 1 ) left uncoated, 2) enterically coated with Eudragit L30D-55 (also referred to as LS30D55 herein) (a coating which should disintegrate at a pH greater than 5.5), or 3) enterically coated with Eudragit FS30D (a coating which should disintegrate at a pH greater than 7). Uncoated capsules containing TSC only (without cyclodextrin) were also administered.

For this study, male Sprague-Dawley (SD) and Wistar rats weighing approximately 300-400g each were fasted for 24 hours prior to each experiment. Water was given ad libitum and coprophagy was prevented by using cages with wire-mesh floors. Anesthesia was induced and maintained with isoflurane. The carotid artery was exposed and cannulated with PE-50 tubing. The cannula was secured using silk sutures and 2% lidocaine was applied to the wound.

After cannulation, a gelatin capsule contain drug product was administered to the stomach via dosing syringe (Torpac, Fairfield NJ) followed by 0.3mL sterile water to the stomach via gavage. Approximately 0.3 mL blood samples were taken for the carotid artery at discrete time intervals following TSC administration. A small amount of sodium heparin was used as an anticoagulant. The blood samples were centrifuged and the resulting plasma volume mixed with 3 volumes of methanol and vortexed. The plasma mixture was then centrifuged and the supernatant was analyzed by HPLC to determine TSC levels.

The graph below shows the concentration in the blood stream after oral administration to rats over a period of 7 hours of dry powders contained in gelatin capsules, with all capsules containing the same amount of TSC (about 4 mg).

TSC only shows the TSC alone in a capsule;

TSC:CD ( 1 :4) uncoated, which is for capsules made of a 1 to 4 mixture of TSC to cyclodextrin but with no coating applied to the gelatin capsules; LS30D55 which is for the same 1 :4 capsule coated with Eudragit LS30D55 (a coating which should disintegrate at a pH greater than 5.5);

FS30D, which is for the same 1 :4 capsule coated with Eudragit FS30D (a coating which should disintegrate at a pH greater than 7).

120 180 240 300

Time following administration, min

The bar graph below shows the percentage improvement in absorption (as compared to TSC only) that is obtained with the uncoated capsules as well as the ones coated with the Eudragits.

Example 7

Neurodegenerative Disease

A major aspect of any neurodegenerative disease is, as stated above, the death of neurons. Studies have been conducted to look at the effects of TSC on neuronal death in different animal models.

The first study in which this was done was one conducted using a rat model of hemorrhagic stroke. For that study, the enzyme collagenase was injected through a burr hole in the skull into the right basal ganglion, which resulted in some of the brain blood vessels rupturing and bleeding out. TSC was then injected starting 3 hours after the collagenase was administered, and the animals were sacrificed 48 hours after the administration of the collagenase.

When there is a hemorrhage in the brain, the blood pools and forms a hematoma.

Around the periphery of this hematoma, there is a death of neuronal cells. However, it was found that treating with TSC resulted in about 20 % less death of the neurons. Use of the stain, fluorojade, allows one to count the numbers of "dead" neurons in a given brain section of the brain. This was done in the area around the hematomas formed in the hemorrhagic stroke rat model. It was found that there was approximately 30% less neuronal death in the animals treated with TSC as shown in the graph below.

Perihematomal Neuronal Cell Death in Rat

Hemorrhagic Stroke Model

Another model that was used examined the effect of TSC on neuronal viability is a rat model of Parkinson's disease. In this model, a burr hole is drilled through the skull and into the region of the substantia nigra. The Parkinson's-like condition is induced by injecting 6- hydroxydopamine (6-OHDA) into the brain. An amount of 10 D g of 6-OHDA is frequently injected in this model, and was used in the first study.

The first study with this model was designed to mimic a study found in the literature, in which pretreatment with crocetin was found to have a beneficial effect on neuronal death following the injection of 10 Dg of 6-OHDA. In the study, TSC was injected at a dosage of 0.1 mg/kg for 7 days preceding the injection of the 6-OHDA (the same dosing regimen as had been done in the crocetin study). Following the injection of the 6-OHDA, no further treatments were given over the next 4 weeks.

At that time the rats were sacrificed and the brains removed and sent to Charles River Laboratories for counts of live neurons. This resulted in counts on the right side of the brain, where the 6-OHDA had been inected, as well as on the untreated left side of the brain. This allowed the comparison of cell death (treated side count/untreated side count) of the controls, which had been pretreated with saline, to the animals pretreated with TSC. The graph below shows these results.

Effect of Pretreatment on Neuronal Death

Parkinson's Disease Model

10 ug

As can be seen, the percentage of live neurons after this treatment is approximately doubled. It should also be noted that this is a very severe model of neuronal death, in that around 85% of the neurons are dead in the controls.

It is of much more interest to know the effect of post-treatment on neuronal death following the injection of 6-OHDA. To investigate that, 5 fJg of 6-OHDA were again injected into the substantia nigra as before, but the treatment began following that injection. The animals were then given daily injections of TSC (0.25 mg/kg) for 4 weeks before sacrifice. The brains were removed and sent to Charles River Laboratories for neuronal counts.

As seen, there are more live neurons (about 20% more) with the TSC treatment, and these data are statistically-significantly different (p < 0.05).

An interesting result of this study was that the brain sections of the non-6-OHDA side of the brains showed almost the same numbers of live neurons regardless of whether saline or TSC were injected, as shown in the graph below. This demonstrates that TSC has no effect on the viability of live neurons.

These results, combined with those found in the hemorrhagic stroke model, teach that TSC exerts a neuroprotective effect in the brain. Example 8

Chemotherapy with TSC

The pulmonary metastasis mouse model is a widely used model for the evaluation of tumor therapy. With B 16 (mouse melanoma) cells, essentially all cells "take" upon

intravenous cell injection in the tail vein, and the tumors are preferentially formed in the lungs. Thus, the term pulmonary metastasis is widely used even though every resulting pulmonary nodule is technically an independent "primary" tumor rather than a true metastasis.

Since the melanin in B 16 cells does not bleach like the rest of the pulmonary tissues, the tumor nodules can be easily visualized after bleaching the extracted lungs in Fekete's solution. There is always a fraction of nodules that is amelanotic ("white") though, and this requires careful counting in order not to underestimate the tumor burden.

For the subject studies, eight-week-old female C57BL/6 mice were obtained from Charles River Laboratories. The mice were housed in groups of 5 or fewer, and received food and water ab libitum.

B l 6 cells were cultured by the Center for Cell Signaling of the University of Virginia using a standard protocol. The cells were received while they were in an exponential growth phase. The cells were supplied at a concentration of 5 X 10 5 cells/mL in Hank's buffered salt solution (HBSS).

The cells were injected immediately upon receipt, and this day was designated as Day 0. On Day 0, all of the mice were injected intravenously in the tail vein with 0. 1 mL of the cell suspension, meaning that each mouse received 0.5 X 10 5 cells. The mice were then left alone until Day 4. On Day 4, the mice were divided into two groups: Group A, consisting of 5 mice, received an intravenous injection of 0.05 mL of saline in the tail vein. Group B, composed of 7 mice, received an intravenous injection of 0.05 mL of a TSC solution in the tail vein, for a TSC dosage of 0.142 mg/kg/day. The same injections were repeated on Days 5 - 8 and on Days 1 1 - 15. On Day 18, the mice were sacrificed using carbon dioxide. The lungs were excised, rinsed and placed in Fekete's Solution and stored at room temperature.

Later, the lungs were assessed visually, in random, blinded order, to obtain a visual count of the numbers of tumors. It was found that there was a mixture of small tumors plus medium-sized and larger tumors. Thus, the tumors were counted visually in two separate groups: small tumors and medium/large tumors. Although the majority of the tumors were black, there were also some white tumors.

The results of the tumor counts are shown in the table and in the following graphs. The table shows the median (med.) numbers of each type of tumor group as well as the mean + standard deviation. The first graph shows the median values and the second graph shows the mean values. The groups were not statistically different due to the large standard deviations.

Table

Small Medium/Large Total

Group N med. mean med. mean med. mean

Saline 5 55 64 + 40 57 60 + 27 109 124 + 66

TSC 7 1 8 32 + 29 19 30 + 24 34 62 + 53 B-16 Tumors in C57B Mice Lungs on Day 18

I Small Tumors

I Medium/Large Tumors

I Total Tumors

Saline TSC

B-16 Tumors in C57B Mice Lungs on Day

■ Small Tumors

■ Medium/Large Tumors

■ Total Tumors

Saline TSC

It will be readily apparent to those skilled in the art that numerous modifications and additions can be made to both the present compounds and compositions, and the related methods without departing from the invention disclosed.