AU2017254872B2 - Enterically coated cysteamine, cystamine and derivatives thereof - Google Patents

Enterically coated cysteamine, cystamine and derivatives thereof Download PDF

Info

Publication number
AU2017254872B2
AU2017254872B2 AU2017254872A AU2017254872A AU2017254872B2 AU 2017254872 B2 AU2017254872 B2 AU 2017254872B2 AU 2017254872 A AU2017254872 A AU 2017254872A AU 2017254872 A AU2017254872 A AU 2017254872A AU 2017254872 B2 AU2017254872 B2 AU 2017254872B2
Authority
AU
Australia
Prior art keywords
cysteamine
composition
cystinosis
cystamine
phthalate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2017254872A
Other versions
AU2017254872A1 (en
Inventor
Ranjan Dohil
Jerry Schneider
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2007210005A external-priority patent/AU2007210005C1/en
Application filed by University of California filed Critical University of California
Priority to AU2017254872A priority Critical patent/AU2017254872B2/en
Publication of AU2017254872A1 publication Critical patent/AU2017254872A1/en
Application granted granted Critical
Publication of AU2017254872B2 publication Critical patent/AU2017254872B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

ENTERICALLY COATED CYSTEAMINE, CYSTAMINE AND DERIVATIVES THEREOF Abstract The disclosure provides oral cysteamine and cystamine formulations useful for treating cystinosis and neurodegenerative diseases and disorders. The formulations provide controlled release compositions that improve quality of life and reduced side-effects.

Description

ENTERICALLY COATED CYSTEAMINE, CYSTAMINE AND DERIVATIVES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority under 35 U.S.C. §119 to U.S. Provisional Application Serial No. 60/762,715, filed January 27, 2006, the disclosure of which is incorporated herein by reference.
[0001a] The present application is a divisional application of Australian Patent Application No. 2016204132 is a divisional application of Australian Patent Application No. 2013267044 , which is a divisional application of Australian Patent Application No. 2007210005 (PCT/US2007/002325). The entire contents of each of which is hereby incorporated by reference.
FIELD OF THE INVENTION [0002] The invention relates to methods, compositions and treatments for metabolic conditions and free radical damage. More specifically, the invention relates to methods and composition useful for treating Cystinosis and neurodegenerative diseases such as Huntington's; Alzheimer's and Parkinson's disease, as free radical and radioprotectants, and as hepto-protectant agents.
BACKGROUND [0003] Cystinosis is a rare, autosomal recessive disease caused by intra-lysosomal accumulation of the amino acid cystine within various tissues, including the spleen, liver, lymph nodes, kidney, bone marrow, and eyes. Nephropathic cystinosis is associated with kidney failure that necessitates kidney transplantation. To date, the only specific treatment for nephropathic cystinosis is the sulfhydryl agent, cysteamine. Cysteamine has been shown to lower intracellular cystine levels, thereby reducing the rate of progression of kidney failure in children, [0004] Cysteamine, through a mechanism of increased gastrin and gastric acid production, is ulcerogenic. When administered orally to children with cystinosis, cysteamine has also been shown to cause a 3-fold increase in gastric acid production, and a 50% rise of serum gastrin levels. As a consequence, subjects that use cysteamine suffer gastrointestinal (Gl) symptoms and are often unable to take cysteamine regularly or at full dose.
13823064
2017254872 30 Apr 2019 [0005] To achieve sustained reduction of leukocyte cystine levels, patients are normally required to take oral cysteamine every 6 hours, which invariably means having to awaken from sleep. However, when a single dose of cysteamine was administered intravenously the leukocyte cystine level remained suppressed for more than 24 hours, possibly because plasma cysteamine concentrations were higher and achieved more rapidly than when the drug is administered orally. Regular intravenous administration of cysteamine would not be practical. Accordingly, there is a need for formulations and delivery methods that would result in higher plasma, and thus intracellular, concentration as well as decrease the number of daily doses and therefore improve the quality of life for patients.
SUMMARY [0006} According to a first aspect, the present invention provides an enterically-coated composition comprising:
a core of cysteamine bitartrate and cystamine or a pharmaceutically-acceptable salt thereof, mixed with one or more pharmaceutically-acceptable binders; and an enteric coating which encases the core, wherein the enteric coating begins to dissolve in an aqueous solution at a pH between about 4.5 to about 5.5.
[0006a] According to a second aspect, the present invention provides a method of treating a subject with nephropathic cystinosis comprising administering a composition according to the first aspect.
[0006b] According to a third aspect, the present invention provides a method of lowering the cystine content of cells in patients having nephropathic cystinosis comprising administering a composition according to the first aspect.
[0006c] According to a fourth aspect, the present invention provides use of a composition according to the first aspect in the manufacture of a medicament for treating a subject in nepropathic cystinosis.
[0006d] According to a fifth aspect, the present invention provides use of a composition according to the first aspect in the manufacture of a medicament for lowering the cystine content of cells in patients having nephropathic cystinosis.
(22619146_1):GGG
2a
2017254872 30 Apr 2019 [0006e] The invention provides a composition comprising an enterically coated cystamine or cystamine derivative.
[0007] The invention also provides a composition comprising an enterically coated cysteamine or cysteamine derivative.
[0008] The invention further provides a composition comprising a coated cystinosis therapeutic agent that has increased uptake in the small intestine compared to a non-coated cystinosis therapeutic agent when administered orally. In one aspect, the coated cystinosis therapeutic agent comprises a cysteamine or cysteamine derivative.
[0009] The invention also provides a method of treating a subject with cystinosis, comprising administering to the subject a composition of the invention.
[0010] The invention also contemplates a method of treating a subject with a neurodegenerative disease or disorder comprising administering to the subject a composition of the invention comprising an enterically coated cystamine or cystamine derivative.
(22619146_1):GGG
2017254872 31 Oct 2017 [0011] The invention provides a pharmaceutical formulation comprising a composition of the invention further including various pharmaceutically acceptable agents (e.g., flavorants, binders and the like) in a pharmaceutically acceptable carrier.
[0012] The invention provides a method of treating cystinosis or a neurodegenerative disease or disorder comprising administering a composition of the invention and a second therapeutic agent.
BRIEF DESCRIPTION OF THE FIGURES [0013] Figure 1 shows enterocolonic tube. (A) Is an abdominal X-ray film showing the radiopaque weighted tip of the tube entering the ascending colon. (B) Is a contrast infused picture. The tube has passed through the small intestine and the tip is confirmed.
[0014] Figure 2 shows mean plasma cysteamine levels taken from patients with cystinosis and control subjects after delivery of drug into various intestinal sites. Error bars are standard error of the mean. In 2 control subjects, most distal point of drug delivery was the mid-ileal region.
[0015] Figure 3 shows the mean change in leukocyte cystine levels, compared with baseline levels, over a 12hour period following delivery of cysteamine into varying intestinal sites. Negative levels signify increased leukocyte cystine depletion compared with baseline.
[0016] Figure 4 shows a scatterplot of plasma cysteamine Cmax vs. AOC of WBC Cystine changes from Baseline. Positive value means decrease from baseline. Negative value means increase from baseline. AOC change from baseline was affected by Cmax for cysteamine (P < .001) .
[0017] Figure 5 shows serial leukocyte cystine levels after drug was given as normal Cystagon and enteric-coated (EC) cysteamine on alternate days. These serial levels were taken during the inpatient phase of the study. Desired
2017254872 30 Apr 2019 cystine levels are below 1 mmol l/2cystine/mg protein. Higher dose enteric-coated (yellow)) drug resulted in prolonged cystine suppression with 12 hour levels still within desired range.
[0018] Figure 6 shows the blood cysteamine levels following a single 450mg dose of Cystagon (series 1), 450mg EC-cysteamine (series 2) and 900mg EC-cysteamine (series 3). The Cmax is higher following EC drug. In addition, the time to Cmax is longer following EC-drug, suggesting that the drug is released from the capsule within the small intestine rather than the stomach.
DETAILED DESCRIPTION [0019] As used herein and in the appended claims, the singular forms a, an, and the include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to a derivative includes a plurality of such derivatives and reference to a subject [0028] In addition, sulfhydryl (SH) compounds such as cysteamine, cystamine, and glutathione are among the most important and active intracellular antioxidants. Cysteamine protects animals against bone marrow and gastrointestinal radiation syndromes. The rationale for the importance of SH compounds is further supported by observations in mitotic cells. These are the most sensitive to radiation injury in terms of cell reproductive death and are noted to have the lowest level of SH compounds. Conversely, S-phase cells, which are the most resistant to radiation injury using the same criteria, have demonstrated the highest levels of inherent SH compounds. In addition, when mitotic cells were treated with cysteamine, they became very resistant to radiation. It has also been noted that cysteamine may directly protect cells against induced mutations. The protection is thought to result from scavenging of free radicals, either directly or via release of protein-bound GSH. An enzyme that liberates cysteamine from coenzyme A has been reported in avian liver and hog kidney. Recently, studies have appeared demonstrating a protective effect of cysteamine against the hepatotoxic agents acetaminophen, includes reference to one or more subjects known to those skilled in the art, and so forth.
(22589317_1):GGG
4a
2017254872 30 Apr 2019 [0020] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice of the disclosed methods and compositions, the exemplary methods, devices and materials are described herein.
[0021] The publications discussed above and throughout the text are provided solely for their disclosure prior to · the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior disclosure.
(22589317_1):GGG
2017254872 31 Oct 2017 [0022] Cystinosis is a metabolic disease characterized by an abnormal accumulation of the amino acid cystine in various organs of the body such as the kidney, eye, muscle, pancreas, and brain. Different organs are affected at different ages.
[0023] There are three clinical forms of cystinosis. Infantile (or nephropathic) cystinosis; late-onset cystinosis; and benign cystinosis. The latter form does not produce kidney damage. Infantile cystinosis is usually diagnosed between 6 and 18 months of age with symptoms of excessive thirst and urination, failure to thrive, rickets, and episodes of dehydration. These findings are caused by a disorder called renal tubulopathy or Fanconi syndrome. As a consequence important nutrients and minerals are lost in the urine. Children with cystinosis also have crystals in their eyes (after one year of age) which may lead to photosensitivity. They also have an increased level of cystine in their white blood cells without adverse effect but allowing the diagnosis to be ascertained. Without specific treatment, children with cystinosis develop endstage renal failure, i.e., lose their kidney function, usually between 6 and 12 years of age. Without cysteamine treatment subjects can develop complications in other organs due to the continued accumulation of cystine throughout the body. These complications can include muscle wasting, difficulty swallowing, diabetes, and hypothyroidism.
[0024] Some symptoms include the inability of the kidneys to concentrate urine and allow important quantities of sodium, potassium, phosphorus, bicarbonate and substances like carnitine to be excreted in the urine. Treatment of symptoms compensates for these urinary losses. Subjects need to drink large quantities of water, because up to 2 to 3 liters of water are lost in the urine every day driving the feeling of being thirsty. In addition, the loss of urinary
2017254872 31 Oct 2017 electrolytes (sodium, potassium, bicarbonate, phosphorus) must be compensated in the subject. It is often necessary to add a salt supplement in the form of sodium chloride. Children also lose bicarbonate and potassium in the urine, which can be compensated for by giving sodium bicarbonate and potassium bicarbonate.
[0025] Specific treatments of cystinosis aim to reduce cystine accumulation within the cells. Cystinosis is currently treated with cysteamine (Cystagon™) . Cysteamine also improves growth of cystinosis children. Cysteamine is only active in a very short period of time not exceeding 5-6 hours, thus requiring administration of Cystagon™ capsules 4 times a day, that is to say about every 6 hours. This treatment is also only effective if continued day after day, indefinitely in order to control the disease. About 1000 children require lifelong treatment to prolong their lives and prevent deterioration of kidney function. However, as mentioned above, cysteamine administration results in increased gastric secretions and is ulcerogenic. In addition, routes and timing of administration provide difficulty for subjects in need of such therapy. Recently, a similar drug called cystamine (the disulfide form of cysteamine) has been studied for neurodegenerative disorders including Huntington's and Parkinson's diseases. Cystamine has similar side-effects and dosing difficulties to that of cysteamine.
[0026] Cysteamine is a potent gastric acid-secretagogue that has been used in laboratory animals to induce duodenal ulceration; studies in humans and animals have shown that cysteamine-induced gastric acid hypersecretion is most likely mediated through hypergastrinemia. In previous studies performed in children with cystinosis who suffered regular upper gastrointestinal symptoms, a single oral dose of cysteamine (11-23 mg/kg) was shown to cause
2017254872 31 Oct 2017 hypergastrinemia and a 2-to 3-fold rise in gastric acidhypersecretion. Symptoms suffered by these individuals included abdominal pain, heartburn, nausea, vomiting, and anorexia. The disclosure demonstrates that cysteamineinduced hypergastrinemia arises, in part, as a local effect on the gastric antral-predominant G-cells in susceptible individuals. The data also suggest that this is also a systemic effect of gastrin release by cysteamine. Depending upon the route of administration, plasma gastrin levels usually peak after intragastric delivery within 30 minutes, whereas the plasma cysteamine levels peak later.
[0027] Subjects with cystinosis are required to ingest oral cysteamine (Cystagon) every 6 hours, day and night. When taken regularly, cysteamine can deplete intracellular cystine by up to 90% (as measured in circulating white blood cells), and this has been’ shown to reduce the rate of progression to kidney failure/transplantation and also to obviate the need for thyroid replacement therapy. Unfortunately, because of the strict treatment regimen and the associated symptoms, nonadherence with cysteamine therapy remains a problem, particularly among adolescent and young adult patients. By reducing the frequency of required cysteamine dosing, adherence to a therapeutic regimen can be improved. The disclosure demonstrates that delivery of cysteamine to the small intestine reduces gastric distress and ulceration and improves bioavailability of cyteamine in the circulation. Delivery of cysteamine into the small intestine is useful due to improved absorption rate from the SI, greater surface area of the SI, and/or less cysteamine undergoing hepatic first pass elimination when absorbed through the small intestine. This disclosure shows a dramatic decrease in leukocyte cystine within an hour of cysteamine delivery.
2017254872 30 Apr 2019 [0028] In addition, sulfhydryl (SH) compounds such as cysteamine, cystamine, and glutathione are among the most important and active intracellular antioxidants. Cysteamine protects animals against bone marrow and gastrointestinal radiation syndromes. The rationale for the importance of SH compounds is further supported by observations in mitotic cells. These are the most sensitive to radiation injury in terms of cell reproductive death and are noted to have the lowest level of SH compounds. Conversely, S-phase cells, which are the most resistant to radiation injury using the same criteria, have demonstrated the highest levels of inherent SH compounds. In addition, when mitotic cells were treated with cysteamine, they became very resistant to radiation. It has also been noted that cysteamine may directly protect cells against induced mutations. The protection is thought to result from scavenging of free radicals, either directly or via release of protein-bound GSH. An enzyme that liberates cysteamine from coenzyme A has been reported in avian liver and hog kidney. Recently, studies have appeared demonstrating a protective effect of cysteamine against the hepatotoxic agents acetaminophen, bromobenzene, and phalloidin.
[0029) Cy stamine, in addition, to its role as a radioprotectant, has been found to alleviate tremors and prolong life gene mutation for Huntington's disease (HD) . The increasing the activity of proteins that protect neurons, from degeneration. Cystamine appears to in mice with the drug may work by nerve cells, or inactivate an enzyme called transglutaminase and thus results in a reduction of huntingtin protein (Nature Medicine 8, 143-149, 2002) . In addition, cystamine was found to increase the levels of certain neuroprotective proteins. However, due to the current methods and formulation of delivery of cystamine, degradation and poor uptake require excessive dosing.
(22589317_1):GGG
2017254872 31 Oct 2017 [0030] The disclosure is not limited with respect to a specific cysteamine or cystamine salt or ester or derivative; the compositions of the disclosure can contain any cysteamine or cystamine, cysteamine or cystamine derivative, or combination of cysteamine or cystamines. The active agents in the composition, i.e., cysteamine or cystamine, may be administered in the form of a pharmacologically acceptable salt, ester, amide, prodrug or analog or as a combination thereof. Salts, esters, amides, prodrugs and analogs of the active agents may be prepared using standard procedures known to those skilled in the art of synthetic organic chemistry and described, for example, by J. March, Advanced Organic Chemistry: Reactions, Mechanisms and Structure, 4th Ed. (New York: WileyInterscience, 1992). For example, basic addition salts are prepared from the neutral drug using conventional means, involving reaction of one or more of the active agent's free hydroxyl groups with a suitable base. Generally, the neutral form of the drug is dissolved in a polar organic solvent such as methanol or ethanol’and the base is added thereto.
The resulting salt either precipitates or may be brought out of solution by addition of a less polar solvent. Suitable bases for forming basic addition salts include, but are not limited to, inorganic bases such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine, or the like. Preparation of esters involves functionalization of hydroxyl groups which may be present within the molecular structure of the drug. The esters are typically acyl-substituted derivatives of free alcohol groups, i.e., moieties which are derived from carboxylic acids of the formula R-COOH where R is alkyl, and typically is lower alkyl. Esters can be reconverted to the free acids, if desired, by using conventional hydrogenolysis or hydrolysis procedures. Preparation of amides and prodrugs
2017254872 31 Oct 2017 can be carried out in an analogous manner. Other derivatives and analogs of the active agents may be prepared using standard techniques known to those skilled in the art of synthetic organic chemistry, or may be deduced by reference to the pertinent literature.
[0031] The disclosure provides delivery methods and compositions that overcome the problems associated with cysteamine and cystamine delivery. The methods of compositions of the disclosure provide enteric-coated compositions that result in less frequent dosing (2X/day vs. 4X/day), increased patient compliance and fewer gastrointestinal side effects (e.g., pain, heartburn, acid production, vomiting) and other side effects (e.g., patients smell like rotten eggs - a particular compliance problem as subjects reach puberty). The disclosure provides entericcoated cysteamine compositions (sulfhydryl/Cystagon™) and cystamine compositions.
[0032] The disclosure provides methods for the treatment of cystinosis, the treatment of neurodegenerative disease such as Alzheimer Disease, Huntington's and Parkinson's disease and free radical damage using enterically coated cysteamine and cystamine, respectively.
[003.3] The disclosure provides composition comprising enterically formulated cysteamine and cystamine derivatives. Examples of cysteamine derivatives include hydrochloride, bitartrate and phosphocysteamine derivatives. Cystamine and cystamine derivatives include sulfated cystamine. Enteric coatings prolong release until the cystamine, cystamine derivative, or cysteamine derivative/Cystagon™ reaches the intestinal tract, typically the small intestine. Because of the enteric coatings, delivery to the small intestine is improved thereby improving uptake of active ingredient while reducing gastric side effects. This will result in a reduction in the need for frequent administration that
2017254872 31 Oct 2017 currently is associated with Cystagon therapy, cystamine and cysteamine therapy.
[0034] An enterically coated drug or tablet refers to a drug or tablet that is coated with a substance--i.e., with an enteric coating--that remains intact in the stomach but dissolves and releases the drug once the small intestine is reached.
[0035] As used herein enteric coating, is a material, a polymer material or materials which encase the medicament core (e.g., cystamine, cysteamine, Cystagon). Typically, a substantial amount or all of the enteric coating material is dissolved before the medicament or therapeutically active agent is released from the dosage form, so as to achieve delayed dissolution of the medicament core. A suitable pHsensitive polymer is one which will dissolve in intestinal juices at a higher pH level (pH greater than 4.5), such as within the small intestine and therefore permit release of the pharmacologically active substance in the regions of the small intestine and not in the upper portion of the GI tract, such as the stomach.
[0036] The coating material is selected such that the therapeutically active agent will be released when the dosage form reaches the small intestine or a region in which the pH is greater than pH 4.5. The coating may be a pHsensitive materials, which remain intact in the lower pH environs of the stomach, but which disintegrate or dissolve at the pH commonly found in the small intestine of the patient. For example, the enteric coating material begins to dissolve in an aqueous solution at pH between about 4.5 to about 5.5. For example, pH-sensitive materials will not undergo significant dissolution until the dosage form has emptied from the stomach. The pH of the small intestine gradually increases from about 4.5 to about 6.5 in the duodenal bulb to about 7.2 in the distal portions of the
2017254872 31 Oct 2017 small intestine (ileum). In order to provide predictable dissolution corresponding to the small intestine transit time of about 3 hours (e.g., 2-3 hours) and permit reproducible release therein, the coating should begin to dissolve within the pH range of the duodenum, and continue to dissolve at the pH range within the small intestine. Therefore, the amount of enteric polymer coating should be sufficient to substantially dissolved during the approximate three hour transit time within the small intestine (e.g., the proximal and mid-small intestine).
[0037] Enteric coatings have been used for many years to arrest the release of the drug from orally ingestible dosage forms. Depending upon the composition and/or thickness, the enteric coatings are resistant to stomach acid for required periods of time before they begin to disintegrate and permit release of the drug in the lower stomach or upper part of the small intestines. Examples of some enteric coatings are disclosed in U.S. Pat. No. 5,225,202 which is incorporated by reference fully herein. As set forth in U.S. Pat. No. 5,225,202, some examples of coating previously employed are beeswax and glyceryl monostearate,- beeswax, shellac and cellulose; and cetyl alcohol, mastic and shellac, as well as shellac and stearic acid (U.S. Pat. No. 2,809,918); polyvinyl acetate and ethyl cellulose (U.S. Pat. No. 3,835,221); and neutral copolymer of polymethacrylic acid esters (Eudragit L30D) (F. W. Goodhart et al., Pharm. Tech., pp. 64-71, April 1984); copolymers of methacrylic acid and methacrylic acid methylester (Eudragits), or a neutral copolymer of polymethacrylic acid esters containing metallic stearates (Mehta et al., U.S. Pat. Nos. 4,728,512 and 4,794,001). Such coatings comprise mixtures of fats and fatty acids, shellac and shellac derivatives and the cellulose acid phthlates, e.g., those having a free carboxyl content. See, Remington's at page 1590, and Zeitova et al.
2017254872 31 Oct 2017 (U.S. Pat. No. 4,432,966), for descriptions of suitable enteric coating compositions. Accordingly, increased adsorption in the small intestine due to enteric coatings of cystamine, cysteamine derivatives (including Cystagon) can result in improvements in cystinosis as well as neurodegenerative diseases including, for example, Huntington's disease.
[0038] Generally, the enteric coating comprises a polymeric material that prevents cysteamine or cystamine release in the low pH environment of the stomach but that ionizes at a slightly higher pH, typically a pH of 4 or 5, and thus dissolves sufficiently in the small intestines to gradually release the active agent therein. Accordingly, among the most effective enteric coating materials are polyacids having a pKa in the range of about 3 to 5. Suitable enteric coating materials include, but are not limited to, polymerized gelatin, shellac, methacrylic acid copolymer type C NF, cellulose butyrate phthalate, cellulose hydrogen phthalate, cellulose proprionate phthalate, polyvinyl acetate phthalate (PVAP), cellulose acetate phthalate (CAP), cellulose acetate trimellitate (CAT), hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate, dioxypropyl methylcellulose succinate, carboxymethyl ethylcellulose (CMEC), hydroxypropyl methylcellulose acetate succinate (HPMCAS), and acrylic acid polymers and copolymers, typically formed from methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate with copolymers of acrylic and methacrylic acid esters (Eudragit NE, Eudragit RL·, Eudragit RS). For example, the enterically coating can comprise Eudragit L3 0D, triethylcitrate, and hydroxypropylmethylcellulose (HPMC), Cystagon® (or other cysteamine derivative), wherein the coating comprises 10 to 13% of the final product.
2017254872 31 Oct 2017 [0039] By pharmaceutically acceptable carrier or pharmaceutically acceptable vehicle are meant materials that are suitable for oral administration and not biologically, or otherwise, undesirable, i.e., that may be administered to a subject along with an active ingredient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of a pharmaceutical composition in which it is contained.
[0040] Similarly, a pharmaceutically acceptable salt, ester or other derivative of an active agent comprise, for example, salts, esters or other derivatives which are not biologically or otherwise undesirable.
[0041] Stabilizing agents refer to compounds that lower the rate at which pharmaceutical degrades, particularly an oral pharmaceutical formulation under environmental conditions of storage.
[0042] By the terms effective amount or therapeutically effective amount of a enteric formulation of cysteamine or cystamine refers to a nontoxic but sufficient amount of the agent to provide the desired therapeutic effect. As will be pointed out below, the exact amount required will vary from subject to subject, depending on the age, weight, and general condition of the subject, the severity of the condition being treated, and the like. An appropriate effective amount in any individual case may be determined by one of ordinary skill in the art using only routine experimentation.
[0043] In one aspect of the disclosure there is provided a stabilized pharmaceutical composition for administration of an cysteamine or cystamine, wherein the cysteamine or cystamine is enterically coated.
[0044] The cysteamine or cystamine is present in the composition in a therapeutically effective amount;
2017254872 31 Oct 2017 typically, the composition is in unit dosage form. The amount of cysteamine or cystamine administered will, of course, be dependent on the age, weight, and general condition of the subject, the severity of the condition being treated, and the judgment of the prescribing physician. Suitable therapeutic amounts will be known to those skilled in the art and/or are described in the pertinent reference texts and literature. In one aspect, the dose is administered twice per day at about 0.5-1.0 g/m2 (e.g., 0.7-0.8 g/m2) body surface area. Current nonenterically coated doses are about 1.35 g/m2 body surface area and are administered 4-5 times per day.
[0045] The entericaly coated cysteamine or cystamine can comprise various excipients, as is well known in the pharmaceutical art, provided such excipients do not exhibit a destabilizing effect on any components in the composition. Thus, excipients such as binders, bulking agents, diluents, disintegrants, lubricants, fillers, carriers, and the like can be combined with the cysteamine or cystamine. For solid compositions, diluents are typically necessary to increase the bulk of a tablet so that a practical size is provided for compression. Suitable diluents include dicalcium phosphate, calcium sulfate, lactose, cellulose, kaolin, mannitol, sodium chloride, dry starch and powdered sugar. Binders are used to impart cohesive qualities to a tablet formulation, and thus ensure that a tablet remains intact after compression. Suitable binder materials include, but are not limited to, starch (including corn starch and pregelatinized starch), gelatin, sugars (including sucrose, glucose, dextrose and lactose), polyethylene glycol, waxes, and natural and synthetic gums, e.g., acacia sodium alginate, polyvinylpyrrolidone, cellulosic polymers (including hydroxypropyl cellulose, hydroxypropyl methylcellulose, methyl cellulose, hydroxyethyl cellulose,
2017254872 31 Oct 2017 and the like), and Veegum. Lubricants are used to facilitate tablet manufacture; examples of suitable lubricants include, for example, magnesium stearate, calcium stearate, and stearic acid, and are typically present at no more than approximately 1 weight percent relative to tablet weight. Disintegrants are used to facilitate tablet disintegration or breakup after administration, and are generally starches, clays, celluloses, algins, gums or crosslinked polymers. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and the like. If desired, flavoring, coloring and/or sweetening agents may be added as well. Other optional components for incorporation into an oral formulation herein include, but are not limited to, preservatives, suspending agents, thickening agents, and the like. Fillers include, for example, insoluble materials such as silicon dioxide, titanium oxide, alumina, talc, kaolin, powdered cellulose, microcrystalline cellulose, and the like, as well as soluble materials such as mannitol, urea, sucrose, lactose, dextrose, sodium chloride, sorbitol, and the like.
[0046] A pharmaceutical composition may also comprise a stabilizing agent such as hydroxypropyl methylcellulose or. polyvinylpyrrolidone, as disclosed in U.S. Pat. No. 4,301,146. Other stabilizing agents include, but are not limited to, cellulosic polymers such as hydroxypropyl cellulose, hydroxyethyl cellulose, methyl cellulose, ethyl cellulose, cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropyl methylcellulose phthalate, microcrystalline cellulose and carboxymethylcellulose sodium; and vinyl polymers and
2017254872 31 Oct 2017 copolymers such as polyvinyl acetate, polyvinylacetate phthalate, vinylacetate crotonic acid copolymer, and ethylene-vinyl acetate copolymers. The stabilizing agent is present in an amount effective to provide the desired stabilizing effect; generally, this means that the ratio of cysteamine or cystamine to the stabilizing agent is at least about 1:500 w/w, more commonly about 1:99 w/w.
[0047] The tablets are manufactured by first enterically coating the cysteamine or cystamine. A method for forming tablets herein is by direct compression of the powders containing the enterically coated cysteamine or cystamine, optionally in combination with diluents, binders, lubricants, disintegrants, colorants, stabilizers or the like. As an alternative to direct compression, compressed tablets can be prepared using wet-granulation or drygranulation processes. Tablets may also be molded rather than compressed, starting with a moist material containing a suitable water-soluble lubricant.
[0048] In an alternative embodiment, the enterically coated cysteamine or cystamine are granulated and the granulation is compressed into a tablet or filled into a capsule. Capsule materials may be either hard or soft, and are typically sealed, such as with gelatin bands or the like. Tablets and capsules for oral use will generally include one or more commonly used excipients as discussed herein.
[0049] For administration of the dosage form, i.e., the tablet or capsule comprising the enterically coated cysteamine or cystamine, a total weight in the range of approximately 100 mg to 1000 mg is used. The dosage form is orally administered to a patient suffering from a condition for which an cysteamine or cystamine would typically be indicated, including, but not limited to, cystinosis and
2017254872 31 Oct 2017 neurodegenerative diseases such as Huntington's, Alzheimer's and Parkinson's disease.
[0050] The compositions of the disclosure can be used in combination with other therapies useful for treating cystinosis and neurodegenerative diseases and disorders. For example, indomethacin therapy (Indocid® or Endol®) is an anti-inflammatory used to treat rheumatoid arthritis and lumbago, but it can be used to reduce water and electrolyte urine loss. In children with cystinosis, indomethacin reduces the urine volume and therefore liquid consumption by about 30%, sometimes by half. In most cases this is associated with an appetite improvement. Indomethacin treatment is generally followed for several years.
[0051] Other therapies can be combined with the methods and compositions of the disclosure to treat diseases and disorders that are attributed or result from cystinosis. Urinary phosphorus loss, for example, entails rickets, and it may be necessary to give a phosphorus supplement. Carnitine is lost in the urine and blood levels are low. Carnitine allows fat to be used by the muscles to provide energy. Hormone supplementation is sometimes necessary. Sometimes the thyroid gland will not produce enough thyroid hormones. This is given as thyroxin (drops or tablets). Insulin treatment is sometimes necessary if diabetes appears, when the pancreas does not produce enough insulin. These treatments have become rarely necessary in children whom are treated with cysteamine, since the treatment protects the thyroid and the pancreas. Some adolescent boys require a testosterone treatment if puberty is late. Growth hormone therapy may be indicated if growth is not sufficient despite a good hydro electrolytes balance. Accordingly, such therapies can be combined with the enterically coated cysteamine and cystamine compositions and methods of the disclosure.
2017254872 31 Oct 2017 [0052] The effectiveness of a method or composition of the disclosure can be assessed by measuring leukocyte cystine concentrations. Dosage adjustment and therapy can be made by a medical specialist depending upon, for example, the severity of cystenosis and/or the concentration of cystine. Additional therapies including the use of omeprazole (Prilosec®) can reduce these symptoms.
[0053] In addition, various prodrugs can be activated by use of the enterically coated cysteamine. Prodrugs are pharmacologically inert, they themselves do not work in the body, but once they have been absorbed, the prodrug decomposes. The prodrug approach has been used successfully in a number of therapeutic areas including antibiotics, antihistamines and ulcer treatments. The advantage of using prodrugs is that the active agent is chemically camouflaged and no active agent is released until the drug has passed out of the gut and into the cells of the body. For example, a number of produgs use S-S bonds. Weak reducing agents, such as cysteamine, reduce these bonds and release the drug. Accordingly, the compositions of the disclosure are useful in combination with pro-drugs for timed release of the drug. In this aspect, a pro-drug can be administered followed by administration of an enterically coated cysteamine compositions of the invention (at a desired time) to activate the pro-drug.
[0054] It is to be understood that while the invention has been described in conjunction with specific embodiments thereof, that the foregoing description as well as the examples which follow are intended to illustrate and not limit the scope of the invention. Other aspects, advantages and modifications within the scope of the invention will be apparent to those skilled in the art to which the invention
EXAMPLES
2017254872 31 Oct 2017 [0055] Subjects. Children with cystinosis, ^12 years old, and taking regular cysteamine bitartrate (Cystagon; Mylan, Morgantown, WV) were recruited to the study (Table I). Adult control patients were recruited locally. Patients with cystinosis had a mean leukocyte cystine level of less than 2.0 nmol half-cystine/mg protein over the past year.
Cysteamine therapy was discontinued 2 days before admission, and acid suppressants, antibiotics, nonsteroidal antiinflammatory drugs, pro-kinetic agents, and antihistamines were discontinued 2 weeks before admission. None of the patients had undergone kidney transplantation. Baseline chemistry, Helicobacter pylori serologic study, complete blood count, and urinalysis were performed.
Table I. Cystinosis patient data
Patient Age (yrs .) Sex Weight (kg) Cys teamine dose (mg)* Serum creatinine (mg/dL)
J 1 IS Male 61.5 500 1.0
2 14 Male 39.4 406 1.2
3 13 Female 39.1 406 1.5
4 19 Female 38.1 406 1.4
5 13 Female 50.1 500 1.0
6 16 Male 58.7 500 3.1
* Dose of cysteamine base delivered into varying delivery sites [0056] Cysteamine bitartrate delivery. Cysteamine was infused through a silicone rubber nasoenteric tube (Dentsleeve Pty Ltd, Australia), 3 mm in diameter and 4.5 meters long. The tube, specifically made for this study, had a tungsten-weighted tip, and immediately proximal to this was an inflatable balloon (5-mL> capacity). Immediately proximal to the balloon was an infusion port (1 mm diameter) through which the drug was delivered. After an overnight fast (except for water), the dose of cysteamine bitartrate (10 mg/kg/dose of base, maximum of 500 mg) was dissolved in 10 mL of water and infused over 1 to 2 minutes. On day 1 of the study, the nasoenteric tube was inserted into the
2017254872 31 Oct 2017 stomach. By day 3 of the study the tube had passed into the proximal small intestine (SI) just distal to the ligament of Treitz (confirmed fluoroscopically). The balloon was then inflated, and peristalsis propelled the tube distally. Tube position within the cecum was confirmed fluoroscopically on day 5 (day 7 in 4 patients because of slow transit). If the tube had migrated too far, it was retracted into the desired location.
[0057] Serum gastrin, cysteamine and leukocyte cystine measurements. After an overnight fast (except for water) blood samples were taken at baseline and at varying intervals after intraluminal delivery of cysteamine. Serum gastrin levels were then measured at 30, 60, 90, and 120 minutes and 3 and 4 hours; cysteamine levels were measured at 0, 5, '10, 20, 30, 45, 60, 75, 90, 105, 120, and 150 minutes and 3, 4, 6, 8, 10, 12, and 16 hours; leukocyte cystine levels were measured at 1, 2, 3, 4, 6, and 12 hours in patients with cystinosis only. Gastrin was measured in picograms/mL· with the Diagnostic Products Corporation (Los Angeles, Calif) gastrin radioimmunoassay-assay kit.
Leukocyte cystine levels were measured in nmol half-cystine per mg protein by the Cystine Determination Lab (La Jolla, Calif).
[0058] To measure plasma cysteamine, 100- pL plasma samples were collected in heparinized vacutainers and spun in a centrifuge within 1 hour, and plasma was stored at -18° C. The concentration of cysteamine was measured by use of tandem mass spectroscopy (API 2000 LC/MS/MS; Applied Biosystems, Foster City, Calif). Cysteamine concentrations were calculated with a calibration curve that was prepared by spiking plasma with buffered cysteamine solutions, and quality control samples were analyzed with each batch.
[0059] Statistical analysis. Mixed model restricted maximum likelihood (REML) repeated measures analysis of
2017254872 31 Oct 2017 variance with subjects as a random effect was performed on the absolute leukocyte cystine levels, on the leukocyte cystine level changes from baseline, and on the area over the curve (AOC) for leukocyte cystine level changes from baseline after cysteamine administration for the subjects with cystinosis. AOC is computationally analogous to area under the curve, but it is applied when values are predominantly decreasing below baseline values. Large AOC values reflect large decreases, and a negative AOC reflects a net increase in value. Main effects for site of delivery, time after delivery, and the interaction between site and time were tested, except just the site effect was tested for AOCs. In the absence of significant interaction when a main effect was detected, Tukey's honestly significant difference test (HSD) was applied to identify where differences occurred within a 5% family wise error rate. The Tukey HSD procedure controls for overall significance level when performing all pairwise comparisons. An additional analysis was performed with plasma cysteamine Cma„ added to the AOC model.
[0060] REML repeated measures analyses of variance with subjects as a random effect were also performed as described above on AUC and the Cmax over time for plasma cysteamine levels separately for the subjects with cystinosis and control subjects and with both subject groups combined. Differences between means for the 3 sites were tested, plus group and group x site interaction effects for the combined groups. If a site effect was detected, Tukey's HSD was applied to determine which sites differed from each other. [0061] REML repeated measures analyses of variance were also performed as described above on gastrin levels. The analyses were performed on 2 versions of datasets: the full dataset and all data after omitting observations collected at 30 minutes (1 subject was missing a blood sample taken at
2017254872 31 Oct 2017 minutes after small intestinal cysteamine delivery). A 5% significance level was used without adjustment for all statistical testing.
[0062] Six patients with cystinosis, (3 male, 3 female) with a mean age of 15.2 years (range 13-19 years) were recruited into the study (Table I). Eight healthy adult control patients (6 male, 2 female) with a mean age of 23.2 years (range 19-28 years) were enrolled. None of the children with cystinosis had undergone kidney transplantation. All control subjects received 500 mg cysteamine base, whereas the mean dose for subjects with cystinosis was 453 mg (range 406-500 mg). All subjects had normal liver function test results. In all subjects the nasoenteric tube passed successfully from the stomach into the upper SI; however, it did not progress any further in 2 subjects with cystinosis. In 2 of the control subjects the tube only reached the mid-ileum but did, however, progress to the cecum in 8 subjects (4 control subjects, 4 with cystinosis). There were no reported adverse effects with the insertion or removal of the nasoenteric tube (Figure 1).
[0063] Symptoms. Only 2 patients (1 male, 1 female) with cystinosis reported regular Gl symptoms before the study, and these had responded to acid-suppression therapy. The male subject had severe retching and emesis about 15 minutes after receiving intragastric cysteamine but did not have any symptoms when the drug was infused into the proximal small intestine. The female child with cystinosis had mild transient nausea after SI drug delivery only. No other symptoms were reported after any other cysteamine delivery in the children with cystinosis. There were no associated adverse events with tube placement or removal.
[0064] Plasma cysteamine. Among the subjects with cystinosis as measured by analysis of variance, the mean plasma cysteamine Cmax and AUCs (of the concentration-time
2017254872 31 Oct 2017 gradient) differed by site of cysteamine delivery (both P < .03). Site (f) refers to either patients with cystinosis or control subjects. For the plasma cysteamine AUCs, the means differed between the duodenal and both gastric and cecal sites of delivery (Tukey HSD global P < .05). Among control subjects, the mean AUG did not differ among delivery sites (P > .4) , but mean Cmax did (P < .05) . For both cystinosis and control groups the mean Cmax values differed only between the duodenum and cecum; mean Cmax values after duodenal versus gastric or gastric versus cecal delivery were not statistically different (Tables II and III) .
Table IX. Mean plasma cysteamine C^x levels (ymol/L) and area under curve (AUC) measurements in cystinosis subjects, controls, and. combined cystinosis and control subjects, after delivery of cysteamine into the stomach, small intestine, and cecum
Cystinosis AUC Cystinosis Control AUC Control ^max Combined AUC Combined
Stomach 35.5 (20.5) 3006 (1112) 39.5 (16.4) 3613 (1384) 37.8 (17.6) 3353 (1267)
Small Intestine 55.8 (13.0) 4299 (1056) 51.1 (20.7) 3988 (1659) 53.2 (17.4) 4047 (1376)
Cecum 21.9 (13.1) 3002 (909) 23.1 (15.3) 2804 (1323) 22.5 (13.2) 2903 (1056)
The standard deviations are in parenthesis
Table III. Comparisons of mean plasma cysteamine Cmax (ymol/L) and AUC measurements for combined cystinosis subjects and control subjects among delivery sites
AUC Qnax
P value* <0.01 <0.01
Stomach vs SI + +
Stomach vs Cecum - -
SI vs Cecum + +
+ Significant difference using Tukey's HSD test (a= 0.05)
- No significant difference * ANOVA test for equality of three delivery sites [0065] When data from the control subjects were combined with cystinosis subject data, there was both a group effect (P < .05) and a site effect (P < .01) for AUCs, with a significant difference between mean AUC levels for the duodenum versus both the stomach and cecum. Cmax values differed among sites (P < .01) but not between groups (P > .4). Group (*)refers to site of intestinal delivery. C^x
2017254872 31 Oct 2017 differed between duodenum versus both stomach and cecum (Figure 2).
[0066] Leukocyte cystine. There were significant differences among the 3 sites of delivery for cystine levels (P < .04), changes from baseline values (P < .0001), and AOCs for changes from baseline (P < .02). A Tukey HSD test, which controls for multiple comparisons, showed that mean leukocyte cystine levels differed between the cecum and stomach sites, but that cecum versus duodenum and stomach versus duodenum produced similar mean values. When the absolute cystine levels or AOCs for changes from baseline levels were evaluated, the significant differences in sites were found between the duodenum and both the stomach and cecum, but not between stomach and cecum (Tukey HSD global P < .05) (Figure 3). Plasma cysteamine Cmax and AUC contributed a statistical effect on AOC (P< .001 and < .02, respectively), even after controlling for delivery site (Figure 4).
[0067] Blood gastrin. For the full gastrin dataset, there was a significant difference among the means for the different delivery sites (P < 0.1), with the cecum resulting in a lower mean from that of the stomach and small intestine. Both group * and site t significant effects were detected after omitting observations from 30 minutes after delivery (P < .05 and P < .01, respectively). The 30-minute observations were omitted because of a missing data set. For these observations, mean levels of gastrin after delivery in the cecum were different from those from both the duodenum and stomach, although the latter did not differ from each other. The 1 boy (14 years) who had severe GI symptoms after intragastric, but not enteric or cecal, cysteamine delivery had a rise in baseline gastrin from 70 pg/mL to 121 pg/mL· at 30 minutes after gastric cysteamine. Within the control group, more than half of the baseline and post-cysteamine
2017254872 31 Oct 2017 gastrin levels remained undetectable (<25 pg/mL), and none of the control subjects had a significant rise in gastrin after cysteamine delivery into any site.
[0068] Patients with cystinosis are required to ingest oral cysteamine (Cystagon) every 6 hours, day and night. When taken regularly, cysteamine can deplete intracellular cystine by up to 90% (as measured in circulating white blood cells), and this has been shown to reduce the rate of progression to kidney failure/transplantation and also to obviate the need for thyroid replacement therapy. Unfortunately, because of the strict treatment regimen and the associated symptoms, nonadherence with cysteamine therapy remains a problem, particularly among adolescent and young adult patients. Certainly, by reducing the frequency of required cysteamine dosing adherence can be improved. The disclosure shows a strong statistical association between the maximum plasma concentration (Cmax) of cysteamine and AOC measurements for leukocyte cystine (P < .001) . A higher Cmax is achieved after delivery of cysteamine into the small intestine than when infused into the stomach or colon; this may be due to improved absorption rate from the SI, greater surface area of the SI, or less cysteamine undergoing hepatic first pass elimination when absorbed rapidly through the small intestine. When data were combined for patients with cystinosis and control subjects, there was a statistical difference between duodenal versus both gastric and colonic delivery for plasma cysteamine Cmax and AUC levels (both P < .05). The lack of similar statistical significance for the cystinosis group alone may simply reflect the small number of patients studied. Changes from baseline leukocyte cystine levels were statistically significant for absolute cystine levels and for AOC when cysteamine was infused into the duodenum compared with both stomach and colon. As shown in Figure 3, the leukocyte
2017254872 31 Oct 2017 cystine levels remained below pre-delivery levels for up to 12 hours after a single dose of cysteamine into the small intestine. This would suggest that effective absorption of cysteamine through the SI, by causing a higher Cmax and AUC on the cysteamine concentration-time gradient, could lead to prolonged depletion of leukocyte cystine and possibly less frequent daily dosing. Another explanation would be that by achieving a high enough plasma cysteamine concentration, more drug reaches the lysosome (where cystine accumulates). In the lysosome the cysteamine reacts with cystine forming the mixed disulfide of cysteamine and cysteine. The mixed disulfide exits the lysosome presumably via the lysine carrier. In the cytosol the mixed disulfide can be reduced by its reaction with glutathione. The cysteine released can be used for protein or glutathione synthesis. The cysteamine released from the mixed disulfide reenters the lysosome where it can react with another cystine molecule. Thus 1 molecule of cysteamine may release many molecules of cystine from the lysosome. This study showed a dramatic decrease in leukocyte cystine within an hour of cysteamine delivery. In retrospect, the finding from this study was that the leukocyte cystine levels remained at the 1-hour level for 24 hours, and even at 48 hours after delivery the levels had not returned to the pre-cysteamine level.
[0069] Cysteamine is a potent gastric acid-secretagogue that has been used in laboratory animals to induce duodenal ulceration; studies in humans and animals have shown that cysteamine-induced gastric acid hypersecretion is most likely mediated through hypergastrinemia. In previous studies performed in children with cystinosis who suffered regular upper gastrointestinal symptoms, a single oral dose of cysteamine (11-23 mg/kg) was shown to cause hypergastrinemia and a 2-to 3-fold rise in gastric acidhypersecretion. Symptoms suffered by these individuals
2017254872 31 Oct 2017 included, abdominal pain, heartburn, nausea, vomiting, and anorexia. Interestingly, only 2 of 6 subjects with cystinosis (who were known to suffer regular cysteamineinduced Gl symptoms) had increased gastrin levels and symptoms, including nausea, retching, and discomfort after intragastric cysteamine. Gastrin levels were only available after small intestinal administration in 1 of the 2 children and the levels remained the same as baseline. Neither child had symptoms after enteric cysteamine delivery. None of the other patients with cystinosis or control subjects had an increase in gastrin levels with cysteamine infused into any site. This would suggest that cysteamine-induced hypergastrinemia may arise as a local effect on the gastric antral-predominant G-cells only in susceptible individuals. In addition, plasma gastrin levels usually peaks after intragastric delivery within 30 minutes, whereas the plasma cysteamine levels peaked later.8,10 In 2 previous studies, children with cystinosis were shown to have a significant rise in plasma gastrin levels after receiving intragastric cysteamine; as part of these study's entry criteria all subjects did, however, suffer with regular Gl symptoms. Data from this study would suggest that cysteamine does not cause hypergastrinemia, and therefore acid-hypersecretion, in all patients with cystinosis. Thus acid suppression therapy would not be recommended in patients with cystinosis without upper Gl symptoms.
[0070] The data suggest that direct administration of cysteamine into the jejunum may result in prolonged leukocyte cystine depletion. In a previous study, a child who had a gastrojejunal feeding tube for oral feeding aversion and severe UGI symptoms, responded to intrajejunal cysteamine with a 3-fold rise in serum gastrin as compared with drug administration into the stomach. The leukocyte cystine response was not measured in this child. Therefore
2017254872 31 Oct 2017 patients with jejunal feeding tubes will have to be further evaluated.
[0071] Figures 5 and 6 shows results from a patient that remained on the twice daily EC-cysteamine for an extended period of time. Over this period the patient's leukocyte cystine levels have been measured regularly. The dose of twice daily EC-cysteamine is titrated against the patients symptoms and cystine levels. The patient's cystine levels have been 0.4, 1.0, 0.35.
[0072] This study provides data that may be used to improve the quality of life for patients with cystinosis. The present formulation of Cystagon comprises cysteamine in a capsule that will dissolve rapidly on contact with water, most likely within the stomach.
[0073] Although a number of embodiments and features have been described above, it will be understood by those skilled in the art that modifications and variations of the described embodiments and features may be made without departing from the teachings of the disclosure or the scope of the invention as defined by the appended claims.
2017254872 31 Oct 2017
Disclosed herein are the following forms of the invention:
1. A composition comprising:
a coated cystinosis therapeutic agent that has increased uptake in the small intestine compared to a noncoated cystinosis therapeutic agent when administered orally.
. The composition of form 1, wherein the coated cystinosis therapeutic agent comprises a cysteamine or cysteamine derivative.
. The composition of form 1, wherein the coated cystinosis therapeutic agent comprises a coating that releases the therapeutic agent when the composition reaches the small intestine or a region of the gastrointestinal tract of a subject in which the pH is greater than pH 4.5.
4. The composition of form 1, wherein the coated cystinosis therapeutic agent comprises a coating selected from the group consisting of polymerized gelatin, shellac, methacrylic acid copolymer type C NF, cellulose butyrate phthalate, cellulose hydrogen phthalate, cellulose proprionate phthalate, polyvinyl acetate phthalate (PVAP), cellulose acetate phthalate (CAP), cellulose acetate trimellitate (CAT), hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate, dioxypropyl methylcellulose succinate, carboxymethyl ethylcellulose (CMEC), hydroxypropyl methylcellulose acetate succinate (HPMCAS), and acrylic acid polymers and copolymers, typically formed from methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate with copolymers of acrylic and methacrylic acid esters.
5. The composition of form 1, wherein the composition is formulated for oral administration.
. The composition of form 1, wherein the composition further comprises a pharmaceutically acceptable carrier.
2017254872 31 Oct 2017
7. The composition of form 1, wherein the composition comprises a stabilizer.
8. A composition comprising:
an enterically coated cystamine or cystamine derivative .
9. The composition of form 8, wherein an enteric coating of the enterically coated cystamine or cystamine derivative releases the cystamine or cystamine derivative when the composition reaches the small intestine or a region of the gastrointestinal tract of a subject in which the pH is greater than pH 4.5.
10. The composition of form 9, wherein the enteric coating is selected from the group consisting of polymerized gelatin, shellac, methacrylic acid copolymer type C NF, cellulose butyrate phthalate, cellulose hydrogen phthalate, cellulose proprionate phthalate, polyvinyl acetate phthalate (PVAP), cellulose acetate phthalate (CAP), cellulose acetate trimellitate (CAT), hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate, dioxypropyl methylcellulose succinate, carboxymethyl ethylcellulose (CMEC), hydroxypropyl methylcellulose acetate succinate (HPMCAS), and acrylic acid polymers and copolymers, typically formed from methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate with copolymers of acrylic and methacrylic acid esters.
11. The composition of form 8, wherein the composition is formulated for oral administration.
12. The composition of form 8, wherein the composition further comprises a pharmaceutically acceptable carrier.
13. The composition of form 8, wherein the composition comprises a stabilizer.
14. A composition comprising:
an enterically coated cysteamine or cysteamine derivative .
2017254872 31 Oct 2017
15. The composition of form 14, wherein an enteric coating of the enterically coated cysteamine or cysteamine derivative releases the cysteamine or cysteamine derivative when the composition reaches the small intestine or a region of the gastrointestinal tract of a subject in which the pH is greater than pH 4.5.
16. The composition of form 15, wherein the enteric coating is selected from the group consisting of polymerized gelatin, shellac, methacrylic acid copolymer type C NF, cellulose butyrate phthalate, cellulose hydrogen phthalate, cellulose proprionate phthalate, polyvinyl acetate phthalate (PVAP), cellulose acetate phthalate (CAP), cellulose acetate trimellitate (CAT), hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate, dioxypropyl methylcellulose succinate, carboxymethyl ethylcellulose (CMEC), hydroxypropyl methylcellulose acetate succinate (HPMCAS), and acrylic acid polymers and copolymers, typically formed from methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate with copolymers of acrylic and methacrylic acid esters.
17. The composition of form 14, wherein the composition is formulated for oral administration.
18. The composition of form 14, wherein the composition further comprises a pharmaceutically acceptable carrier.
19. The composition of form 14, wherein the composition comprises a stabilizer.
20. A method of treating a subject with cystinosis, comprising administering to the subject a composition of form 1 or 14.
21. A method of treating a subject with a neurodegenerative disease or disorder comprising administering to the subject a composition of form 8.
2017254872 31 Oct 2017
22. A pharmaceutical formulation comprising a composition of form 1, 8 or 14 in a pharmaceutically acceptable carrier.
23. A method of form 20 or 21,, further comprising treating the subject with a second therapeutic agent.
24. A method comprising:
administering a prodrug;
administering a composition of form 14 under conditions and in an amount effective to activate the prodrug.

Claims (16)

  1. CLAIMS :
    1. An enterically-coated composition comprising;
    a core of cysteamine bitartrate and cystamine or a pharmaceutically-acceptable salt thereof, mixed with one or more pharmaceutically-acceptable binders; and an enteric coating which encases the core, wherein the enteric coating begins to dissolve in an aqueous solution at a pH between about 4.5 to about 5.5.
  2. 2. The composition of claim 1, wherein the composition is in the form of a tablet or granules.
  3. 3. The composition of claim 2, wherein the granules are loaded into a capsule.
  4. 4. The composition of claim 1, wherein the enteric coating comprises a compound selected from the group consisting of polymerized gelatin, shellac, methacrylic acid copolymer type CNF, cellulose butyrate phthalate, cellulose hydrogen phthalate, cellulose propionate phthalate, polyvinyl acetate phthalate (PVAP), cellulose acetate phthalate (CAP), cellulose acetate trimellitate (CAT), hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate, dioxypropyl methylcellulose succinate, carboxymethyl ethylcellulose (CMEC), hydroxypropyl methylcellulose acetate succinate (HPMCAS), and acrylic acid polymers and copolymers formed from methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate with copolymers of acrylic and methacrylic acid esters.
  5. 5. The composition of claim 4, wherein the enteric coating comprises a methacrylic acid ester.
  6. 6. The composition of claim 1, wherein the composition is formulated for oral administration.
    (22583998_1):GGG
    2017254872 15 May 2019
  7. 7. A method of treating a subject with nephropathic cystinosis comprising administering a composition of any one of claims 1-6 twice per day.
  8. 8. The method of claim 7, wherein each administration comprises an amount of cysteamine bitartrate that would yield a dose of 100 to 1000 mg cysteamine free base.
  9. 9. The method of claim 7, wherein administration maintains white blood-cell cystine concentration at less than 1 nmol ^ystine/mg protein.
  10. 10. The method of claim 8, wherein the total daily dose of cysteamine free base is about 1.35 g/m2 body surface area.
  11. 11. A method of lowering the cystine content of cells in patients having nephropathic cystinosis comprising administering a composition of any one of claims 1-6.
  12. 12. The method of claim 11, wherein each administration comprises an amount of cysteamine bitartrate that would yield a dose of 100 to 1000 mg cysteamine free base.
  13. 13. The method of claim 11, wherein administration maintains white blood-cell cysteine concentration at less than 1 nmol ^ystine/mg protein.
  14. 14. The method of claim 12, wherein the total daily dose of cysteamine free base is about 1.35 g/m2 body surface area.
  15. 15. Use of a composition of any one of claims 1-6 in the manufacture of a medicament for treating a suject with nephropathic cystinosis.
    (22583998_1):GGG
    2017254872 15 May 2019
  16. 16. Use of a composition of any one of claims 1-6 in the manufacture of a medicament for lowering the cystine content of cells in patients having nephropathic cystinosis.
AU2017254872A 2006-01-27 2017-10-31 Enterically coated cysteamine, cystamine and derivatives thereof Active AU2017254872B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2017254872A AU2017254872B2 (en) 2006-01-27 2017-10-31 Enterically coated cysteamine, cystamine and derivatives thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US60/762,715 2006-01-27
AU2007210005A AU2007210005C1 (en) 2006-01-27 2007-01-26 Enterically coated cysteamine, cystamine and derivatives thereof
AU2013267044A AU2013267044B2 (en) 2006-01-27 2013-12-05 Enterically coated cysteamine, cystamine and derivatives thereof
AU2016204132A AU2016204132B2 (en) 2006-01-27 2016-06-17 Enterically coated cysteamine, cystamine and derivatives thereof
AU2017254872A AU2017254872B2 (en) 2006-01-27 2017-10-31 Enterically coated cysteamine, cystamine and derivatives thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2016204132A Division AU2016204132B2 (en) 2006-01-27 2016-06-17 Enterically coated cysteamine, cystamine and derivatives thereof

Publications (2)

Publication Number Publication Date
AU2017254872A1 AU2017254872A1 (en) 2017-11-16
AU2017254872B2 true AU2017254872B2 (en) 2019-06-06

Family

ID=49883371

Family Applications (3)

Application Number Title Priority Date Filing Date
AU2013267044A Active AU2013267044B2 (en) 2006-01-27 2013-12-05 Enterically coated cysteamine, cystamine and derivatives thereof
AU2016204132A Active AU2016204132B2 (en) 2006-01-27 2016-06-17 Enterically coated cysteamine, cystamine and derivatives thereof
AU2017254872A Active AU2017254872B2 (en) 2006-01-27 2017-10-31 Enterically coated cysteamine, cystamine and derivatives thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
AU2013267044A Active AU2013267044B2 (en) 2006-01-27 2013-12-05 Enterically coated cysteamine, cystamine and derivatives thereof
AU2016204132A Active AU2016204132B2 (en) 2006-01-27 2016-06-17 Enterically coated cysteamine, cystamine and derivatives thereof

Country Status (1)

Country Link
AU (3) AU2013267044B2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050008702A1 (en) * 2003-05-22 2005-01-13 Joaquina Faour Rupturing controlled release device having a preformed passageway
US20050137125A1 (en) * 2003-12-19 2005-06-23 Chan Bill P. Compositions and methods for treating diabetes

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2386817B (en) * 2002-02-20 2006-08-23 Walcom Animal Science Feed for fish and use therof
CN101822658A (en) * 2004-05-03 2010-09-08 奥加生物药业(I.P.3)有限公司 Cysteamine is used for the treatment of hypercholesterolemia and diabetic complication

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050008702A1 (en) * 2003-05-22 2005-01-13 Joaquina Faour Rupturing controlled release device having a preformed passageway
US20050137125A1 (en) * 2003-12-19 2005-06-23 Chan Bill P. Compositions and methods for treating diabetes

Also Published As

Publication number Publication date
AU2016204132A1 (en) 2016-07-14
AU2016204132B2 (en) 2017-08-03
AU2013267044B2 (en) 2016-07-07
AU2017254872A1 (en) 2017-11-16
AU2013267044A1 (en) 2014-01-09

Similar Documents

Publication Publication Date Title
US11311507B2 (en) Enterically coated cysteamine, cystamine and derivatives thereof
AU2017254872B2 (en) Enterically coated cysteamine, cystamine and derivatives thereof

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)