US20060100278A1 - Dosage forms and related therapies - Google Patents

Dosage forms and related therapies Download PDF

Info

Publication number
US20060100278A1
US20060100278A1 US10/525,345 US52534505A US2006100278A1 US 20060100278 A1 US20060100278 A1 US 20060100278A1 US 52534505 A US52534505 A US 52534505A US 2006100278 A1 US2006100278 A1 US 2006100278A1
Authority
US
United States
Prior art keywords
alkyl
trientine
composition
drug
diabetic
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.)
Abandoned
Application number
US10/525,345
Other languages
English (en)
Inventor
Garth Cooper
John Baker
Nigel Beelev
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.)
PHILERA NEW ZEALAND Ltd
Original Assignee
Protemix Corp Ltd
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
Application filed by Protemix Corp Ltd filed Critical Protemix Corp Ltd
Assigned to PROTEMIX CORPORATION LIMITED reassignment PROTEMIX CORPORATION LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEELEY, NIGEL ROBERT ARNOLD, BAKER, JOHN RICHARD, COOPER, GARTH JAMES SMITH
Publication of US20060100278A1 publication Critical patent/US20060100278A1/en
Assigned to PHILERA NEW ZEALAND LIMITED reassignment PHILERA NEW ZEALAND LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PROTEMIX CORPORATION LIMITED
Priority to US14/573,211 priority Critical patent/US9339479B2/en
Priority to US15/096,570 priority patent/US9993443B2/en
Priority to US15/966,433 priority patent/US10543178B2/en
Priority to US16/719,404 priority patent/US11419831B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/132Amines having two or more amino groups, e.g. spermidine, putrescine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/131Amines acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/30Copper compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/04Chelating agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • the subject invention pertains to doses and dosage forms of therapeutic agents and their use in methods for the treatment, reversal or amelioration of diseases, disorders and/or conditions in a mammal (hereafter “treating”).
  • Mammals that may be treated using the described and claimed doses and dosage forms include, for example, a human being having, or at risk for developing, microvascular and/or macrovascular damage, for example, cardiovascular tissue damage and, in particular, mammals including human beings that have or are at risk for developing undesired copper levels, including copper levels that can cause or lead to tissue damage, including but not limited to vessel damage.
  • Treatment includes but is not limited to therapies to ameliorate and/or reverse, in whole or in part, damage resulting from diseases, disorders or conditions that are characterized in any part by copper-involved or mediated damage of tissue and/or vasculature, and/or to copper-involved or mediated impairment of normal tissue stem cell responses.
  • the invention has application inter alia, for example, to diabetes-related and non-diabetes-related heart failure, macrovascular disease or damage, microvascular disease or damage, and/or toxic (e.g., hypertensive) tissue and/or organ disease or damage (including such ailments as may, for example, be characterized by heart failure, cardiomyopathy, myocardial infarction, and related arterial and organ diseases) by administration of an active copper-chelating compound such as, for example, one or more of trientine, salts of trientine, prodrugs of trientine and salts of such prodrugs, analogs of trientine and salts and prodrugs of such analogs, and/or active metabolites of trientine and salts and prodrugs of such metabolites, including but not limited to N-acetyl trientine and salts and prodrugs of N-acetyl trientine.
  • an active copper-chelating compound such as, for example, one or more of trientine, salts of trientine, prodrugs of
  • Diabetes mellitus is a chronic condition characterized by the presence of fasting hyperglycemia and the development of widespread premature atherosclerosis. Patients with diabetes have increased morbidity and mortality due to cardiovascular diseases, especially coronary artery disease. Vascular complications in diabetes may be classified as microvascular, affecting the retina, kidney and nerves and macrovascular, predominantly affecting for example coronary, cerebrovascular and peripheral arterial circulation.
  • the chronic hyperglycemia of diabetes is associated with long-term damage, dysfunction, and failure of various organs, especially the eyes, kidneys, nerves, heart, and blood vessels and long-term complications of diabetes include retinopathy with potential loss of vision; nephropathy leading to renal failure; peripheral neuropathy with risk of foot ulcers, amputation, and Charcot joints; and autonomic neuropathy causing gastrointestinal, genitourinary, and cardiovascular symptoms and sexual dysfunction.
  • Glycation of tissue proteins and other macromolecules and excess production of polyol compounds from glucose are among the mechanisms thought to produce tissue damage from chronic hyperglycemia.
  • Diabetic patients have an increased incidence of atherosclerotic cardiovascular, peripheral vascular, and cerebrovascular disease. Hypertension, abnormalities of lipoprotein metabolism, and periodontal disease are also found in people with diabetes.
  • Hyperglycemia induces a large number of alterations in vascular tissue that potentially promote accelerated atherosclerosis.
  • vascular tissue that potentially promote accelerated atherosclerosis.
  • oxidative stress and protein kinase C (PKC) activation.
  • PKC protein kinase C
  • Atherosclerosis accounts for virtually 80% of all deaths among North American diabetic patients, compared with one-third of all deaths in the general North American population, and more than 75% of all hospitalizations for diabetic complications are attributable to cardiovascular disease.
  • American Diabetes Association “Consensus statement: role of cardiovascular risk factors in prevention and treatment of macrovascular disease in diabetes,” Diabetes Care 16:72-78 (1993).
  • CAD is the leading cause of death in people with type 2 diabetes, regardless of duration of diabetes.
  • Stamler I., et al. “Diabetes, other risk factors, and 12-yr cardiovascular mortality for men screened in the Multiple Risk Factor Intervention Trial,” Diabetes Care 16:434-444 (1993); Donahue R. P., & Orchard T. J., “Diabetes mellitus and macrovascular complications. An epidemiological perspective,” Diabetes Care 15:1141-1155 (1992).
  • the increased cardiovascular risk is said to be particularly striking in women.
  • Barrett Connor E. L., et al. “Why is diabetes mellitus a stronger risk factor for fatal ischemic heart disease in women than in men?
  • CAD is not confined to particular forms of diabetes, however, and is prevalent in both type 1 and type 2 diabetes. In type 1 diabetes, an excess of cardiovascular mortality is generally observed after the age of 30. Krolewski A. S., et al., “Magnitude and determinants of coronary artery disease in juvenile-onset, insulin-dependent diabetes mellitus,” Am J Cardiol 59:750-75 5 (1987). CAD risk was reported in this study to increase rapidly after age 40, and by age 55, 35% of men and women with type 1 diabetes die of CAD, a rate of CAD mortality that far exceeded that observed in an age-matched nondiabetic cohort. Id.
  • Diabetic nephropathy in type 1 diabetics also increases the prevalence of CAD. Nephropathy leads to accelerated accumulation of AGEs in the circulation and tissue and parallels the severity of renal functional impairment. Makita Z., et al., “Advanced glycosylation end products in patients with diabetic nephropathy,” N Engl J Med 325:836-842 (1991). In diabetic patients reaching end-stage renal disease, overall mortality has been reported to be greater than in nondiabetic patients with end-stage renal disease. The relative risk for age-specific death rate from myocardial infarction among all diabetic patients during the first year of dialysis is reportedly 89-fold higher than that of the general population.
  • Diabetic heart disease is further characterized by more severe CAD at a younger age, a 4-fold increase in frequency of heart failure, post-acute myocardial infarction and a disproportionate increase in left ventricular hypertrophy.
  • Subjects with type 2 diabetes also manifest a disproportionate increase in mortality within the first 24-hours post-acute myocardial infarction. Acute intervention can ameliorate this risk.
  • Malmberg K. Br Med J 314:1512-5 (1997).
  • PCT Application No. PCT/NZ99/00161 (published as WO00/18392 on 6 Apr. 2000) relates to methods of treating a mammalian subject predisposed to and/or suffering from diabetes mellitus with a view to minimizing the consequences of macrovascular and microvascular damage to the patent which comprises, in addition to any treatment in order to control blood glucose levels, at least periodically controlling copper, for example, in the subject.
  • An assay method is disclosed in PCT Application No. PCT/NZ99/00160 (published as WO00/18891 on 6 Apr. 2000).
  • a range of different treatment agents are disclosed in PCT/NZ99/00161. These included copper chelating agents.
  • Metals are present naturally in the body and many are essential for cells (e.g., Cu, Fe, Mn, Ni, Zn). However, all metals are toxic at high concentrations. One reason metals may become toxic relates to their ability to cause oxidative stress, particularly redox active transition metals, which can take up or give off an electron (e.g., Fe2+/3+, Cu+/2 ⁇ ) that can give rise to free radicals that cause damage (Jones et al., “Evidence for the generation of hydroxyl radicals from a chromium (V) intermediate isolated from the reaction of chromate with glutathione,” Biochim. Biophys. Acta 286:652-655 (1991); Li, Y. & Trush, M.
  • V chromium
  • Cardiac function is commonly assessed by measuring the ejection fraction.
  • a normal left ventricle ejects at least 50% of its end-diastolic volume each beat.
  • a patient with systolic heart failure commonly has a left ventricular ejection fraction less than 30% with a compensatory increase in end-diastolic volume.
  • Hemodynamic studies conducted on diabetic subjects without overt congestive heart failure have observed normal left ventricular systolic function (LV ejection fraction) but abnormal diastolic function suggesting impaired left ventricular relaxation or filling. See Regan, et al., J. Gun. Invest. 60:885-99 (1977).
  • Diagnosis maybe made, for example, by non-invasive measurements.
  • mitral diastolic blood flow measured by Doppler echocardiography is a direct measure of left ventricular filling. The most commonly used measurement is the AlE ratio. Normal early diastolic filling is rapid and is characterized by an E-wave velocity of around 1 m/sec.
  • Late diastolic filling due to atrial contraction is only a minor component, and the A-wave velocity is perhaps around 0.5 m/sec. This gives a normal AIE ratio of approximately 0.5. With diastolic dysfunction, early diastolic filling is impaired, atrial contraction increases to compensate, and the AlE ratio increases to more than 2.0.
  • inotropic drugs are designed to improve the contraction of the failing heart.
  • a heart with pure diastolic dysfunction is already contracting normally and it is believed that inotropic drugs will increase the risk of arrhythmias.
  • vasodilator drugs that reduce after-load and improve the emptying of the ventricle because ejection fraction and end-diastolic volume are already normal. After-load reduction may even worsen cardiac function by creating a degree of outflow obstruction.
  • Diuretics are the mainstay of therapy for heart failure by controlling salt and water retention and reducing filling pressures. However, they are contraindicated in diastolic dysfunction where compromised cardiac pump function is dependent on high filling pressures to maintain cardiac output.
  • Venodilator drugs such as the nitrates, which are very effective in the management of systolic heart failure by reducing pre-load and filling pressures, are understood to be poorly tolerated by patients with diastolic heart failure. Ejection fraction and end-systolic volume are often normal and any reduction in pre-load leads to a marked fall in cardiac output.
  • beta-blockers in heart failure because of their potential to worsen pump function. There is also concern regarding the administration of beta-blockers to patients with diabetes who are treated with sulphonylurea drugs and insulin due to a heightened risk of severe hypoglycaemia.
  • compositions capable of addressing damage arising from disease states, disorders or conditions of the cardiovascular tree (including the heart) and dependent organs (e.g., retina, kidney, nerves, etc.) that involve, concern or relate to, for example, elevated or undesired copper levels such as elevated non-intracellular free copper values levels.
  • the described and claimed therapies also provide low dose controlled release and/or low dose extended release compositions useful for the reversal and/or amelioration of structural damage in a subject whether diabetic or not, having copper levels capable of diminishment in order to treat, for example, the heart, the macrovasculature, the microvasculature, and/or long-term complications of diabetes, including cardiac structure damage.
  • Cardiac structure damage includes, but is not limited to, for example, atrophy, loss of myocytes, expansion of the extracellular space and increased deposition of extracellular matrix (and its consequences) and/or coronary artery structure damage selected from at least media damage (the muscle layer) and intima damage (the endothelial layer) (and its consequences), systolic function, diastolic function, contractility, recoil characteristics and ejection fraction.
  • Diseases, disorders and conditions relating to the cardiovascular tree and/or dependent organs that may be treated by the methods and compositions of the present invention include, for example, any one or more of (1) disorders of the heart muscle (cardiomyopathy or myocarditis) such as idiopathic cardiomyopathy, metabolic cardiomyopathy which includes diabetic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy; (2) atheromatous disorders of the major blood vessels (macrovascular disease) such as the aorta, the coronary arteries, the carotid arteries, the cerebrovascular arteries, the renal arteries, the iliac arteries, the femoral arteries, and the popliteal arteries; (3) toxic, drug-induced, and metabolic (including hypertensive and/or diabetic disorders of small blood vessels (microvascular disease) such as the retinal arterioles, the glomerular arterioles, the vasa nervorum, cardiac arterioles, and associated capillary
  • the present invention is based, in part, on new doses and dosage forms for treatments aimed at reduction in available free copper that are useful, for example, in treating and preventing macrovascular, microvascular and/or toxic/metabolic diseases of the kind referenced herein and in tissue repair processes. This is irrespective of the glucose metabolism of the subject and irrespective of whether or not fructosamine oxidase is involved in any such disease.
  • the invention also relates to doses and dosage forms of treatments relating to the cardiovascular accumulation of redox-active transition metal ions in diabetes.
  • Conditions occurring in the context of diabetes and/or impaired glucose tolerance in which the suppression of normal stem cell responses can cause impairment of normal tissue responses, and that would be improved with therapy to lower copper values using the doses and dosage forms of treatments described herein, include the following:
  • Heart failure A significant regeneration of cardiac tissues can occur within a few days of cardiac transplantation. The likely mechanism is migration of stem cells from extra-cardiac sites to the heart, with subsequent differentiation of such cells into various specialized cardiac cells, including myocardial, endothelial and coronary vascular cells. We have determined that copper accumulation in cardiac tissues is likely to severely impair these regenerative responses and that, for example, there is a role for acute intravenous therapy with a copper chelator in the treatment of heart failure, including but not limited to, diabetic heart failure.
  • AMI Acute Myocardial infarction
  • AMI is accompanied by proliferation of cells in the ventricular myocardium when, for example, AMI occurs in the context of diabetes.
  • the presence of elevated tissue levels of redox-active transition metals suppresses normal stem cell responses, resulting in impaired structural and functional repair of damaged tissues.
  • the mechanism of the impairment of cardiac function in, for example, diabetes is believed to be a toxic effect of accumulated transition metals on tissue dynamics, resulting in impaired tissue regeneration caused in turn by suppression of normal stem cell responses, which mediate physiological tissue regeneration by migration to damaged tissue from external sites.
  • Treatment of AMI for example, in the context of diabetes, will be improved by acute (if necessary, parenteral) as well as by subsequent chronic administration of a copper chelator as described herein.
  • Tissue damage resulting from infection Processes of normal tissue repair following infection require intervention of mobilized stem cells that migrate to sites of tissue damage to effect tissue regeneration and repair of, for example, the various layers of blood vessels. Such tissue damage repair will be impaired by suppressed stem cell responses, such as those caused by the build up of redox-active transition metals (particularly copper) in tissues, for examples the walls of blood vessels. Tissue damage repair, including repair following infection, will be improved, for example, in people with diabetes by use of the doses and dosage forms of treatments described herein.
  • Diabetic kidney damage Treatment of diabetics and others having kidney failure by administration of a copper chelator according to the doses and dosage forms of treatments described herein will improve organ regeneration by restoring normal tissue healing by allowing stem cells to migrate and differentiate normally.
  • this invention features a method of diminishment of available free copper values in any at risk subject, whether diabetic or not, and particularly a subject not suffering from Wilson's Disease and who has copper levels capable of diminishment by the administration of an effective amount of an agent capable of lowering copper levels in a subject.
  • a preferred copper chelator is trientine, including trientine acid addition salts and active metabolites including, for example, N-acetyl trientine, and analogs, derivatives, and prodrugs thereof.
  • Alternative names for trientine include N,N′-Bis(2-aminoethyl)-1,2-ethanedi-amine; triethylenetetramine (“TETA”); 1,8-diamino-3,6-diazaoctane; 3,6-diazaoctane-1,8-diamine; 1,4,7,10-tetraazadecane; trien; TECZA; and, triene.
  • the trientine is rendered less basic (e.g., as a acid addition salt).
  • trientine is modified, i.e., it may be as an analogue or derivative of trientine (or an analogue or derivative of a copper-chelating metabolite of trientine, for example, N-acetyl trientine).
  • Derivatives of trientine or trientine salts or analogues include those modified with polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the structure of PEG is HO—(—CH 2 —CH 2 —O—) n —H. It is a linear or branched, neutral polyether available in a variety of molecular weights.
  • Analogues of trientine include, for example, compounds in which one or more sulfur molecules is substituted for one or more of the NH groups in trientine.
  • analogues include, for example, compounds in which trientine has been modified to include one or more additional —CH 2 groups.
  • the chemical formula of trientine is NH 2 —CH 2 —CH 2 —NH—CH 2 —CH 2 —NH—CH 2 —CH 2 —NH 2 .
  • the empirical formula is C 6 N 4 H 18 .
  • Analogues of trientine include, for example: 1. SH-CH 2 -CH 2 -NH-CH 2 -CH 2 -NH-CH 2 -CH 2 -NH 2 , 2. SH-CH 2 -CH 2 -S-CH 2 -CH 2 -NH-CH 2 -CH 2 -NH 2 , 3.
  • One or more hydroxyl groups may also be substituted for one or more amine groups to create an analogue of trientine (with or without the substitution of one or more sulfurs for one or more nitrogens). Additional analogues, including acyclic and cyclic analogues, are provided below in reference to Formula I and Formula II.
  • trientine is delivered as a prodrug of trientine or a copper chelating metabolite of trientine.
  • Salts of trientine include, in one embodiment, acid addition salts such as, for example, those of suitable mineral or organic acids.
  • Salts of trientine (such as acid addition salts, e.g., trientine dihydrochloride) act as copper-chelating agents that aid in the elimination of copper from the body by forming a stable soluble complex that is readily excreted by the kidney.
  • the present invention consists in a method of (1) improvement or reversal, in whole or in part, of at least one or more of cardiac structure damage in the subject (for example, atrophy, loss of myocytes, expansion of the extracellular space, and/or increased deposition of extracellular matrix (and its consequences), and/or, (2) improvement, in whole or in part, of any one or more of systolic function, diastolic function, contractility, recoil characteristics, and ejection fraction (as determined, for example, by ultrasound, MRI or other imaging), and/or (3) improvement or reversal, in whole or in part, of any damage from disorders of the heart muscle, macrovascular disease, microvascular disease, and/or plaque rupture of athereomatous lesions of major blood vessels (and the consequences thereof), and/or (4) Improvement or reversal, in whole or in part, of damage resulting from diabetic kidney disease, diabetic nephropathy, copper accumulation in the kidney, and/or damage to the renal arteries.
  • This method may comprise:
  • composition is provided to the subject in a dosage form(s) capable of providing a lower effective dose, and a less pulsile exposure to trientine than has hitherto been the case with “QID” Wilson's disease regimens.
  • the present invention consists in a method of ameliorating or reversing, in whole or in part, in (I) a diabetic human being or other diabetic mammal or (II) a human being or other mammal with copper levels capable of diminishment (“the subject”) one or more of atrophy, loss of myocytes, expansion of the extracellular space, and/or increased deposition of extracellular matrix (and its consequences) and/or coronary artery structure damage, including media damage (the muscle layer) and intima damage (the endothelial layer) (and its consequences).
  • the method comprises or includes the step of administration and/or self administration to the subject a slow or sustained release dosage form sufficient to provide effective chelation of copper for an overall diminishment thereof in the subject, said dosage form having as the or an active agent trientine, at least one salt of trientine, at least one trientine prodrug or a salt of such a prodrug, at least one trientine analog or a salt or prodrug of such an analog, and/or at least one active metabolite of trientine or a salt or prodrug of such a metabolite, including but not limited to N-acetyl trientine and salts and prodrugs of N-acetyl trientine (“trientine active agents”).
  • the subject has been identified prior to treatment as being at risk.
  • the present invention consists in a method of ameliorating or reversing, in whole or in part, any one or more of systolic dysfunction, diastolic dysfunction, contractility, lack of desired recoil characteristics and/or desired ejection fraction function (as determined, for example, by ultrasound, MRI or other imaging), disorders of the heart muscle, macrovascular disease, microvascular disease and plaque rupture of athereomatous lesions of major blood vessels (and consequences thereof), in a subject at risk who is either (I) a diabetic subject or (II) a subject with copper levels capable of diminishment, said method comprising the step of administration and/or self administration of a low, slow, and/or controlled release dosage form sufficient to provide effective treatment, for example, by chelation of copper, for an overall diminishment thereof in the subject, said dosage form having one or more copper chelators, for example, one or more trientine active agents.
  • Diseases, disorders and conditions that are usefully be targeted by the compositions and procedures of the present invention include, but are not limited to, any one or more of the following: diabetic cardiomyopathy, diabetic acute coronary syndrome (e.g.; myocardial infarction (MI), diabetic hypertensive cardiomyopathy, acute coronary syndrome associated with impaired glucose tolerance (IGT), acute coronary syndrome associated with impaired fasting glucose (IFG), hypertensive cardiomyopathy associated with IGT, hypertensive cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with IGT, ischemic cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with coronary heart disease (CHD), acute coronary syndrome not associated with any abnormality of the glucose metabolism, hypertensive cardiomyopathy not associated with any abnormality of the glucose metabolism, ischemic cardiomyopathy not associated with any abnormality of the glucose metabolism (irrespective of whether or not such ischemic cardiomyopathy is associated with coronary heart disease or not), and any one or more disease of the vascular tree including, by way of example
  • the present invention consists in the use of at least one trientine active agent together with other material(s) appropriate for the dosage form, in the manufacture of a sustained release dosage form useful for ameliorating or reversing, in whole or in part, in a subject who is either (I) a diabetic subject or (II) a subject with copper levels capable of diminishment, damage associated with, or irregularity of, any one or more of systolic function, diastolic function, contractility, recoil characteristics and ejection fraction (e.g., as determined clinically, by ultrasound, MRI or other imaging), and/or any one or more of at least some of any damage arising from diabetic kidney disease, diabetic nephropathy and/or copper accumulation in the kidney and/or at least some of any damage to the renal arteries, and/or_cardiac structure damage selected from one or more of atrophy, loss of myocytes, expansion of the extracellular space and increased deposition of extracellular matrix (and its consequences), and/or coronary artery
  • the present invention in another aspect provides a method for treating a subject having, for example, any one or more of the indications as defined herein comprising the parenteral administration of a composition having a therapeutically effective amount of a copper chelator wherein said therapeutically effective amount administered is from about 5 mg to about 1100 mg per does and/or per day.
  • the copper chelator is a trientine active agent.
  • Trientine active agents include, for example, salt(s) of trientine, a trientine prodrug or a salt of such a prodrug, a trientine analogue or a salt or prodrug of such an analog, and/or at least one active metabolite of trientine or a salt or prodrug of such a metabolite, including but not limited to N-acetyl trientine and salts and prodrugs of N-acetyl trientine.
  • Trientine active agents also include the analogues of Formulae I and II.
  • trientine active agents including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on, for example, include from 10 mg to 1100 mg, 10 mg to 1000 mg, 10 mg to 900 mg, 20 mg to 800 mg, 30 mg to 700 mg, 40 mg to 600 mg, 50 mg to 500 mg, 50 mg to 450 mg, from 50-100 mg to about 400 mg, 50-100 mg to about 300 mg, 110 to 290 mg, 120 to 280 mg, 130 to 270 mg, 140 to 260 mg, 150 to 250 mg, 160 to 240 mg, 170 to 230 mg, 180 to 220 mg, 190 to 210 mg, and/or any other amount within the ranges as set forth.
  • composition may include, depending on the rate of parenteral administration, for example, solutions, suspensions, emulsions that can be administered by subcutaneous, intravenous, intramuscular, intradermal, intrastemal injection or infusion techniques.
  • the formulation can further include, for example, any one or more of the following a buffer, for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5, a carbohydrate or polyhydric alcohol tonicifier, an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • a buffer for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5
  • a carbohydrate or polyhydric alcohol tonicifier an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • the formulation of the invention should be substantially isotonic.
  • An isotonic solution may be defined as a solution that has a concentration of electrolytes, non-electrolytes, or a combination of the two that will exert an equivalent osmotic pressure as that into which it is being introduced, in this case, mammalian tissue.
  • substantially isotonic is meant within ⁇ 20% of isotonicity, preferably within ⁇ 10%.
  • the formulated product may be included within a container, typically, for example, a vial, cartridge, prefilled syringe or disposable pen.
  • the present invention provides a parenteral composition comprising a therapeutically effective amount of a copper chelator to be administered to a subject having any one or more of the indications as defined herein.
  • the indications include, for example, diabetic cardiomyopathy, diabetic acute coronary syndrome (e.g.; myocardial infarction—MI), diabetic hypertensive cardiomyopathy, acute coronary syndrome associated with impaired glucose tolerance (IGT), acute coronary syndrome associated with impaired fasting glucose (IFG), hypertensive cardiomyopathy associated with IGT, hypertensive cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with IGT, ischemic cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with coronary heart disease (CHD), disorders of the heart muscle (cardiomyopathy or myocarditis) that include, for example, idiopathic cardiomyopathy, metabolic cardiomyopathy which includes diabetic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy, acute coronary syndrome not associated with any abnormality of glucose metabolism, hypertensive cardiomyopathy not associated with any abnormality of glucose metabolism, ischemic cardiomyopathy not associated with any abnormality of glucose metabolism
  • the copper chelator is a trientine active agent
  • Trientine active agents include, for example, salt(s) of trientine, a trientine prodrug or a salt of such a prodrug, a trientine analog or a salt or prodrug of such an analog, and/or at least one active metabolite of trientine or a salt or prodrug of such a metabolite, including but not limited to N-acetyl trientine and salts and prodrugs of N-acetyl trientine.
  • a therapeutically effective amount of a copper chelator for example, one or more trientine active agents, including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on, is from about 5 mg to 1200 mg per day.
  • Other therapeutically effective dose ranges include from 10 mg to 1100 mg, 10 mg to 1000 mg, 10 mg to 900 mg, 20 mg to 800 mg, 30 mg to 700 mg, 40 mg to 600 mg, 50 mg to 500 mg, 50 mg to 450 mg, from 50-100 mg to about 400 mg, 50-100 mg to about 300 mg, 110 to 290 mg, 120 to 280 mg, 130 to 270 mg, 140 to 260 mg, 150 to 250 mg, 160 to 240 mg, 170 to 230 mg, 180 to 220 mg, 190 to 210 mg, and/or any other amount within the ranges as set forth.
  • composition may include, depending on the rate of parenteral administration, for example, solutions, suspensions, emulsions that can be administered by subcutaneous, intravenous, intramuscular, intradermal, intrastemal injection or infusion techniques.
  • the formulation can further include, for example, any one or more of the following a buffer, for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5, a carbohydrate or polyhydric alcohol tonicifier, an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • a buffer for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5
  • a carbohydrate or polyhydric alcohol tonicifier an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • the formulation of the invention should be substantially isotonic.
  • An isotonic solution may be defined as a solution that has a concentration of electrolytes, non-electrolytes, or a combination of the two that will exert an equivalent osmotic pressure as that into which it is being introduced, in this case, mammalian tissue.
  • substantially isotonic is meant within ⁇ 20% of isotonicity, preferably within ⁇ 10%.
  • the formulated product may be included within a container, typically, for example, a vial, cartridge, prefilled syringe or disposable pen.
  • the present invention provides the use of a therapeutically effective amount of a copper chelator in the manufacture of a medicament for the treatment of a subject having any one or more of the following indications: diabetic cardiomyopathy, diabetic acute coronary syndrome (e.g.; myocardial infarction—MI), diabetic hypertensive cardiomyopathy, acute coronary syndrome associated with impaired glucose tolerance (IGT), acute coronary syndrome associated with impaired fasting glucose (IFG), hypertensive cardiomyopathy associated with IGT, hypertensive cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with IGT, ischemic cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with coronary heart disease (CHD), disorders of the heart muscle (cardiomyopathy or myocarditis) that include, for example, idiopathic cardiomyopathy, metabolic cardiomyopathy which includes diabetic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy, acute coronary syndrome not
  • the copper chelator is a trientine active agent
  • Trientine active agents include, for example, salt(s) of trientine, a trientine prodrug or a salt of such a prodrug, a trientine analog or a salt or prodrug of such an analog, and/or at least one active metabolite of trientine or a salt or prodrug of such a metabolite, including but not limited to N-acetyl trientine and salts and prodrugs of N-acetyl trientine.
  • the therapeutically effective amount of a copper chelator for example, a trientine active agents, including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on, is from about 5 mg to 1200 mg per day.
  • a trientine active agents including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on.
  • Other therapeutically effective dose ranges include from 10 mg to 1100 mg, 10 mg to 1000 mg, 10 mg to 900 mg, 20 mg to 800 mg, 30 mg to 700 mg, 40 mg to 600 mg, 50 mg to 500 mg, 50 mg to 450 mg, from 50-100 mg to about 400 mg, 50-100 mg to about 300 mg, 110 to 290 mg, 120 to 280 mg, 130 to 270 mg, 140 to 260 mg, 150 to 250 mg, 160 to 240 mg, 170 to 230 mg, 180 to 220 mg, 190 to 210 mg, and/or any other amount within the ranges as set forth.
  • composition may include, depending on the rate of parenteral administration, for example, solutions, suspensions, emulsions that can be administered by subcutaneous, intravenous, intramuscular, intradermal, intrastemal injection or infusion techniques.
  • the formulation can further include, for example, any one or more of the following a buffer, for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5, a carbohydrate or polyhydric alcohol tonicifier, an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • a buffer for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5
  • a carbohydrate or polyhydric alcohol tonicifier an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • the formulation of the invention should be substantially isotonic.
  • An isotonic solution may be defined as a solution that has a concentration of electrolytes, non-electrolytes, or a combination of the two that will exert an equivalent osmotic pressure as that into which it is being introduced, in this case, mammalian tissue.
  • substantially isotonic is meant within ⁇ 20% of isotonicity, preferably within ⁇ 10%.
  • the formulated product may be included within a container, typically, for example, a vial, cartridge, prefilled syringe or disposable pen.
  • parenteral administration includes, but is not limited, to any one or more of the following administration routes; subcutaneous, intravenous, intramuscular, intraperitoneal, intrasternal, intraarticular or intrastemal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or vaginally.
  • administration routes include, but is not limited, to any one or more of the following administration routes; subcutaneous, intravenous, intramuscular, intraperitoneal, intrasternal, intraarticular or intrastemal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or vaginally.
  • Urine must be collected in copper-free glassware. It is expected that the patient probably will be in the desired state of negative copper balance if 0.5 to 1.0 milligram of copper is present in a 24-hour collection of urine.
  • the present invention in one aspect provides a method for treating a subject having, for example, any one or more of the indications as defined herein comprising the parenteral administration of a composition having a therapeutically effective amount of a copper chelator wherein said therapeutically effective amount administered parenterally per dose rate is in the range of about 0.1 mg/kg to about 40 mg/kg based on the body weight of the subject.
  • the therapeutically effective amount of copper chelator for example, one or more trientine active agents, including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on, is from about 5 mg to 1200 mg per day.
  • Other therapeutically effective dose ranges include from 10 mg to 1100 mg, 10 mg to 1000 mg, 10 mg to 900 mg, 20 mg to 800 mg, 30 mg to 700 mg, 40 mg to 600 mg, 50 mg to 500 mg, 50 mg to 450 mg, from 50-100 mg to about 400 mg, 50-100 mg to about 300 mg, 110 to 290 mg, 120 to 280 mg, 130 to 270 mg, 140 to 260 mg, 150 to 250 mg, 160 to 240 mg, 170 to 230 mg, 180 to 220 mg, 190 to 210 mg, and/or any other amount within the ranges as set forth.
  • composition may include, depending on the rate of parenteral administration, for example, solutions, suspensions, emulsions that can be administered by subcutaneous, intravenous, intramuscular, intradermal, intrastemal injection or infusion techniques.
  • the formulation can further include, for example, any one or more of the following a buffer, for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5, a carbohydrate or polyhydric alcohol tonicifier, an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • a buffer for example, an acetate, phosphate, citrate or glutamate buffer to obtain a pH of the final formulation from approximately 5.0 to 9.5
  • a carbohydrate or polyhydric alcohol tonicifier an antimicrobial preservative that may be selected from the group of, for example, m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol and a stabilizer.
  • the formulation of the invention should be substantially isotonic.
  • An isotonic solution may be defined as a solution that has a concentration of electrolytes, non-electrolytes, or a combination of the two that will exert an equivalent osmotic pressure as that into which it is being introduced, in this case, mammalian tissue.
  • substantially isotonic is meant within ⁇ 20% of isotonicity, preferably within ⁇ 10%.
  • the formulated product may be included within a container, typically, for example, a vial, cartridge, prefilled syringe or disposable pen.
  • the present invention consists in a transdermal patch, pad, wrap or bandage (“patch”) capable of being adhered or otherwise associated with the skin of a subject, said patch being capable of delivering an effective amount of one or more trientine active agents when so applied to a subject who is either (I) a diabetic subject or (II) a subject with copper levels capable of diminishment to ameliorate or reverse, in whole or in part, any one or more of systolic dysfunction, diastolic dysfunction, contractility dysfunction, recoil dysfunction and ejection fraction dysfunction (as determined, for example, by ultrasound, MRI or other imaging) and/or any one or more of at least some of any damage arising from diabetic kidney disease, diabetic nephropathy and/or copper accumulation in the kidney and/or at least some of any damage to the renal arteries and/or cardiac structure damage selected from one or more of atrophy, loss of myocytes, expansion of the extracellular space and increased deposition of extracellular matrix (and its consequences), and/or coronary
  • the present invention consists in an article of manufacturing comprising a vessel containing as a CR, SR and/or ER dosage form or one or more active agents, or containing in CR, SR and/or ER dosage forms one or more pharmaceutically copper chelators, including but not limited to one or more acceptable trientine active agents; and instructions for use for ameliorating and/or reversing, in whole or in part, in subject who is either (I) a diabetic subject or (II) a subject with copper levels capable of diminishment any one or more of the above-listed indications.
  • the present invention consists in an article of manufacture comprising packaging material; and contained within the packaging material one or more pharmaceutically acceptable trientine active agents in a CR, SR and/or ER dosage form, wherein the packaging material has a label that indicates that the dosage form can be used for ameliorating, reversing and/or improving in a subject who is either (I) a diabetic subject or (II) a subject with copper levels capable of diminishment, any one or more of the above-listed indications.
  • the dosage form, effective amount and/or dosage regimen as herein referred to is able to provide an effective daily dosage to the subject of a trientine active agent (when expressed, for example, as the dihydrochloride salt of trientine, irrespective of whether or not the dosage unit includes that salt) of 4 g per day or below although if given orally the dosage is from 1 mg to 4 g per day.
  • a trientine active agent when expressed, for example, as the dihydrochloride salt of trientine, irrespective of whether or not the dosage unit includes that salt
  • the oral dose delivery (cumulative or otherwise) is in the range of from 200 mg to 4 g per day if given orally. In a further embodiment the daily dosage is such as to deliver 1.2 g per day or below.
  • the dosage delivery is to provide, for example, when expressed as trientine dihydrochloride or other compound herein, a delivery into the subject (irrespective of the dosage included in the dosage unit or units) being administered of from 1 mg to 1.2 g per day. If orally administered the dosage is from 200 mg to 1.2 g per day.
  • the dosage is such as to deliver, for example, the trientine active agent in a dosage unit that administers the trientine active agent at a pH of from 7.2 to 7.6 (preferably a pH of 7.4 ⁇ 0.1).
  • the dosage of, for example, the trientine active agent, for example, trientine dihydrochloride in sustained release is such that there is always less of the active ingredient in a subject's body than results from the 250 mg plus oral dosage forms for Wilson's disease.
  • a sustained release dosage form or forms of, for example, the trientine active agent, for example, trientine dihydrochloride is provided that are suitable for once daily administration and that provide sustained or controlled and long-lasting in vivo release.
  • the form may deliver, for example, not more than 10% trientine dihydrochloride in about 5 hours at an acid pH of about ⁇ 4.5 and delivers greater than 50% of trientine dihydrochloride in 12 hrs at a pH of about ⁇ 6.5 in a controlled manner during in vivo and in vitro dissolution.
  • the present invention provides a method of administering an effective amount of, for example, one or more trientine active agents formulated in a delayed release preparation (DR), a slow release preparation (SR), an extended release preparation (ER), a controlled release preparation (CR) and/or in a repeat action preparation (RA).
  • DR delayed release preparation
  • SR slow release preparation
  • ER extended release preparation
  • CR controlled release preparation
  • RA repeat action preparation
  • the formulations of DR, SR, ER, RA, or CR are suitable for use in the treatment of any of the indications listed herein, including but not limited to, heart failure, diabetic heart disease, acute coronary syndrome, hypertensive heart disease, ischemic heart disease, coronary artery disease, peripheral arterial disease, Wilson's disease, or any form of cancer.
  • Formulations of DR, SR, ER, RA, or CR may contain an effective dosage unit for delivery to the subject of from about 1 mg to abut 600 mg per unit of at-least one trientine active agent, although in a further embodiment the total daily dose rate is from between 5 grams to 1 mg and may work to maintain a desired blood plasma concentration of the trientine active agent for a desired period of time, preferably at least about from between 18 to 24 hours.
  • the present invention consists in a formulation of, for example, at least one trientine active agent that maintains constant plasma concentrations of the trientine active agent for extended periods and is effective in removing copper from the body of subjects with any one or more of the indications listed herein, including but not limited to, heart failure, diabetic heart disease, acute coronary syndrome, hypertensive heart disease, ischemic heart disease, coronary artery disease, peripheral arterial disease, Wilson's disease, or any form of cancer.
  • a device containing, for example, one or more trientine active agents in a monolithic matrix device and employed for the treatment of any one or more of the indications listed herein, including but not limited to, heart failure, diabetic heart disease, acute coronary syndrome, hypertensive heart disease, ischemic heart disease, coronary artery disease, peripheral arterial disease, Wilson's disease, or any form of cancer.
  • the monolithic matrix device contains said one or more trientine active agents in a dispersed soluble matrix, in which said one or more trientine active agents becomes increasingly available as the matrix dissolves or swells.
  • the monolithic matrix device may include but is not limited to one or more of the following excipients: hydroxypropylcellulose (BP) or hydroxypropyl cellulose (USP); hydroxypropyl methylcellulose (BP, USP); methylcellulose (BP, USP); calcium carboxymethylcellulose (BP, USP); acrylic acid polymer or carboxy polymethylene (Carbopol) or Carbomer (BP, USP); or linear glycuronan polymers such as alginic acid (BP, USP), for example those formulated into microparticles from alginic acid (alginate)-gelatin hydrocolloid coacervate systems, or those in which liposomes have been encapsulated by coatings of alginic acid with poly-L-lysine membranes.
  • BP
  • the monolithic matrix contains, for example, said one or more trientine active agents particles in a lipid matrix or insoluble polymer matrix, including but not limited to preparations formed from Carnauba wax (BP; USP); medium-chain triglyceride such as fractionated coconut oil (BP) or triglycerida saturata media (PhEur); or cellulose ethyl ether or ethylcellulose (BP, USP).
  • BP Carnauba wax
  • medium-chain triglyceride such as fractionated coconut oil (BP) or triglycerida saturata media (PhEur); or cellulose ethyl ether or ethylcellulose (BP, USP).
  • the lipids can be present in said monolithic matrix from between 20-40% hydrophobic solids w/w. The lipids may remain intact during the release process.
  • the device contains in addition to, for example, said one or more trientine active agents, one or more of the following, for example: a channeling agent, such as sodium chloride or one or more sugars, which leaches from the formulation, forming aqueous micro-channels (capillaries) through which solvent enters, and through which drug is released.
  • a channeling agent such as sodium chloride or one or more sugars, which leaches from the formulation, forming aqueous micro-channels (capillaries) through which solvent enters, and through which drug is released.
  • the device is any hydrophilic polymer matrix, in which said one or more, for example, trientine active agents is/are compressed as a mixture with any water-swellable hydrophilic polymer.
  • the trientine active agent(s), for example, contained in the hydrophilic polymer matrix may be between 20-80% (w/w).
  • the hydrophilic polymer matrix contains in addition to said one or more, for example, trientine active agents any one or more of the following, for example: a gel modifier such as one or more of a sugar, counter ions, a pH buffer, a surfactant, a lubricant such as a magnesium stearate and/or a glidant such as colloidal silicon dioxide.
  • a gel modifier such as one or more of a sugar, counter ions, a pH buffer, a surfactant, a lubricant such as a magnesium stearate and/or a glidant such as colloidal silicon dioxide.
  • the present invention consists in any device containing an effective amount of, for example, said one or more tritentine active agents comprising or including a rate-controlling membrane surrounding a drug reservoir and containing lactulose mixed with microcrystalline cellulose.
  • the ratio of lactulose to microcrystalline cellulose may be, for example, about 60:40.
  • a dose rate of 1.2 g per day is capable of being provided by the use of capsules of 300 mg trientine hydrochloride given half an hour before meals two being given in the morning and two being given at night.
  • a measurement of free copper [which equals total plasma copper minus ceruloplasmin-bound copper] can be made using the procedure disclosed in the Merck & Co Inc datasheet (www.Merck.com) for SYPRINE® (trientine dihydrochloride) capsules: It states, “The most reliable index for monitoring treatment is the determination of free cooper in the serum, which equals the difference between quantitatively determined total copper and ceruloplasmin-copper. Adequately treated subjects will usually have less than 10 mcg free copper/dL of serum. Therapy may be monitored with a 24-hour urinary copper analysis periodically. Urine must be collected in copper-free glassware. Since a low copper diet should keep copper absorption down to less than one milligram a day, the subject probably will be in the desired state of negative copper balance if 0.5 to 1.0 milligram of copper is present in a 24-hour collection of urine”.
  • FIG. 1 shows the urine excretion in diabetic and non-diabetic animals in response to increasing doses of trientine or equivalent volume of saline, wherein urine excretion in diabetic and nondiabetic animals in response to increasing doses of trientine (bottom; 0.1, 1.0, 10, 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush injected at time shown by arrow) or an equivalent volume of saline (top), and each point represents a 15 min urine collection period (see Example 2 Methods for details); error bars show SEM and P values are stated if significant (P ⁇ 0.05).
  • FIG. 2 shows urine excretion in non-diabetic and diabetic animals receiving increasing doses of trientine or an equivalent volume of saline, wherein urine excretion in diabetic (top) and nondiabetic (bottom) rats receiving increasing doses of trientine (0.1, 1.0, 10, 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush injected at time shown by arrow) or an equivalent volume of saline, and each point represents a 15 min urine collection period (see Example 2 Methods for details); error bars show SEM and P values are stated if significant (P ⁇ 0.05).
  • FIG. 3 shows copper excretion in the urine of diabetic and non-diabetic animals receiving increasing doses of trientine or an equivalent volume of saline, wherein copper excretion in urine of diabetic (top) and nondiabetic (bottom) rats receiving increasing doses of trientine (0.1, 1.0, 10, 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush injected at time shown by arrow) or an equivalent volume of saline, and each point represents a 15 min urine collection period (see Example 2 methods for details); error bars show SEM and P values are stated if significant (P ⁇ 0.05).
  • FIG. 4 shows the same information in FIG. 18 with presentation of urinary copper excretion per gram of bodyweight, wherein urinary copper excretion per gram of bodyweight in diabetic and nondiabetic animals in response to increasing doses of trientine (bottom; 0.1, 1.0, 10, 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush injected at time shown by arrow) or an equivalent volume of saline (top), and each point represents a 15 min urine collection period (see Example 2 methods for details); error bars show SEM and P values are stated if significant (P ⁇ 0.05).
  • FIG. 7 shows the iron excretion in urine of diabetic and non-diabetic animals receiving increasing doses of trientine or an equivalent volume of saline, wherein iron excretion in urine of diabetic (top) and nondiabetic (bottom) rats receiving increasing doses of trientine (0.1, 1.0, 10, 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush injected at time shown by arrow) or an equivalent volume of saline, and each point represents a 15 min urine collection period (see Example 2 methods for details); error bars show SEM and P values are stated if significant (P ⁇ 0.05).
  • FIG. 8 shows the urinary iron excretion per gram of bodyweight in diabetic and non-diabetic animals receiving trientine or saline, wherein urinary iron excretion per gram of bodyweight in diabetic and nondiabetic animals in response to increasing doses of trientine (bottom; 0.1, 1.0, 10, 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush injected at time shown by arrow) or an equivalent volume of saline (top), and each point represents a 15 min urine collection period (see Example 2 methods for details); error bars show SEM and P values are stated if significant (P ⁇ 0.05).
  • FIG. 11 shows urinary [Cu] by AAS ( ⁇ ) and EPR ( ⁇ ) following sequential 10 mg.kg ⁇ 1 (A) and 100 (B) trientine boluses, as in FIG. 19 ; (inset) background-corrected EPR signal from 75-min urine indicating presence of Cu II -trientine; *, P ⁇ 0.05, **, P ⁇ 0.01 vs. control.
  • FIG. 12 is a table comparing the copper and iron excretion in the animals receiving trientine or saline, which is a statistical analysis using a mixed linear model.
  • FIG. 13 shows the body weight of animals changing over the time period of experiment in Example 5.
  • FIG. 14 shows the glucose levels of animals changing over the time period of the experiment in Example 5.
  • FIG. 15 is a diagram showing cardiac output in animals as measured in Example 5.
  • FIG. 16 is a diagram showing coronary flow in animals as measured in Example 5.
  • FIG. 17 is a diagram showing coronary flows normalized to final cardiac weight in animals as measured in Example 5./
  • FIG. 18 is a diagram showing aortic flow in animals as measured in Example 5.
  • FIG. 19 is a diagram showing the maximum rate of positive change in pressure development in the ventricle with each cardiac cycle (contraction) in animals as measured in Example 5.
  • FIG. 20 is a diagram showing the maximum rate of decrease in pressure in the ventricle with each cardiac cycle (relaxation) in animals as measured in Example 5.
  • FIG. 21 shows the percentage of functional surviving hearts at each after-load in animals as measured in Example 5.
  • FIG. 22 shows the structure of LV-myocardium from STZ-diabetic and matched non-diabetic control rats following 7-w oral trientine treatment, wherein cardiac sections were cut following functional studies. Each image is representative of 5 independent sections per heart ⁇ 3 hearts per treatment.
  • a Untreated-control
  • b Untreated-diabetic
  • c Trientine treated diabetic
  • d Trientine-treated non-diabetic control.
  • FIG. 24 shows a randomized, double blind, placebo-controlled trial comparing effects of oral trientine and placebo on urinary Cu excretion from male humans with uncomplicated T2DM and matched non-diabetic controls, wherein urinary Cu excretion ( ⁇ mol.2 h ⁇ 1 on day 1 (baseline) and day 7 following a single 2.4-g oral dose of trientine or matched placebo to subjects described in Table 9, placebo-treated T2DM, ⁇ , placebo-treated control, •, trientine-treated T2DM, ⁇ ; trientine treated control, ⁇ .
  • Cu excretion from T2DM following trientine-treatment was significantly greater than that from trientine-treated non-diabetic controls (P ⁇ 0.05).
  • FIG. 25 shows mean arterial pressure (MAP) response in diabetic and nondiabetic animals to 10 mg.kg ⁇ 1 Trientine in 75 ⁇ l+125 ⁇ l saline flush (or an equivalent volume of saline). Each point represents one minute averages of data points collected every 2 seconds. The time of drug (or saline) administration is indicated by the arrow. Error bars show SEM, and
  • FIG. 26 shows the ultraviolet-visible spectral trace of the trientine containing formulation after being stored for 15 days and upon the addition of copper to form the trientine-copper complex.
  • the traces were taken on day 0 (control formulation) and day 15.
  • day 0 control formulation
  • day 15 There were three formulations containing trientine one was stored in the dark at 4° C., the second at room temperature (21° C.) in the dark and a third at room temperature in daylight. When the spectral was taken copper was added.
  • a reduction in extra-cellular copper values will be advantageous in providing a reduction in and/or a reversal of copper-associated damage, for example, in whole or in part, as well as improved tissue repair by restoration of normal tissue stem cell responses.
  • trientine dihydrochloride The half-life of various copper chelators, for example, trientine, indicated for the treatment and reversal of heart failure and coronary heart disease, is relatively short—being approximately 2 hours.
  • trientine should be taken in addition to current therapies, at a maximum tolerated dose, utilizing a dose regimen that fits its pharmacokinetic and site of action profiles.
  • the plasma concentration of trientine after oral administration to a patient see Miyazaki, K., et al., “Determination of trientine in plasma of subjects with high-performance liquid chromatography,” Chem Pharm Bull 38:1035-38 (1998).
  • Subjects with heart failure and/or coronary artery disease are frequently on multiple drug regimens. Improved copper chelator doses, dose preparation, and/or routes of administration for said doses and dose preparations is needed for this reason as well.
  • the invention is related to and provides novel doses and dose formulations, and routes of administration of various doses and dose formulations, of copper chelators such as, for example, trientine active agents.
  • Trientine active agents include, for example, trientine, salts of trientine, prodrugs of trientine and salts of such prodrugs, analogs of trientine and salts and prodrugs of such analogs, and/or active metabolites of trientine and salts and prodrugs of such metabolites, including but not limited to N-acetyl trientine and salts and prodrugs of N-acetyl trientine.
  • Wilson's disease is due to an inherited defect in copper excretion into the bile by the liver. The resulting copper accumulation and copper toxicity results in liver disease, and in some patients, brain damage. Patients present, generally between the ages of 10 and 40 years, with liver disease, neurological disease of a movement disorder type, or behavioral abnormalities, and often with a combination of these. Wilson's disease is effectively treated with orally administered copper chelators. It has been demonstrated that chelated copper in patients with Wilson's disease is excreted primarily through the feces, either by the effective chelation of copper in the gut (or inhibition of absorption), or by partial restoration of mechanisms that allow for excretion of excess copper via urine or into the bile, or a combination of the two. See Siegemund R, et al., “Mode of action of triethylenetetramine dihydrochloride on copper metabolism in Wilson's disease,” Acta Neurol Scand. 83(6):364-6 (June 1991).
  • the compounds may also be formulated for parenteral injection (including, for example, by bolus injection or continuous infusion) and may be presented in unit dose form in ampules, pre-filled syringes, small bolus infusion containers, or in multi-does containers with an added preservative.
  • doses and dose formulations of copper chelators that maintain desired blood and tissue levels may be prepared that are highly effective in causing removal of systemic copper from the body via the urine and at lower doses than required for oral administration given that gut copper need not be excreted, and will be more effective in the treatment of any condition in which pathologically increased or undesired tissue copper plays a role in disease initiation or progression.
  • diseases include any of the indications identified herein, including but not limited to the following: heart failure, diabetic heart disease, acute coronary syndrome, hypertensive heart disease, ischemic heart disease, coronary artery disease, peripheral arterial disease, and forms of cancer amenable to treatment by copper chelation.
  • Trientine is a strongly basic moiety with multiple nitrogens that can be converted into a large number of suitable associated acid addition salts using an acid, for example, by reaction of stoichiometrically equivalent amounts of trientine and of the acid in an inert solvent such as ethanol or water and subsequent evaporation if the dosage form is best formulated from a dry salt. Possible acids for this reaction are in particular those that yield physiologically acceptable salts.
  • Nitrogen-containing copper chelators for example, trientine active agents such as, for example, trientine, that can be delivered as a salt(s) (such as acid addition salts, e.g., trientine dihydrochloride) act as copper-chelating agents, which aids the elimination of copper from the body by forming a stable soluble complex that is readily excreted by the kidney.
  • trientine active agents such as, for example, trientine
  • inorganic acids can be used, e.g., sulfuric acid, nitric acid, hydrohalic acids such as hydrochloric acid or hydrobromic acid, phosphoric acids such as orthophosphoric acid, sulfamic acid. This is not an exhaustive list.
  • organic acids can be used to prepare suitable salt forms, in particular aliphatic, alicyclic, araliphatic, aromatic or heterocyclic mono-or polybasic carboxylic, sulfonic or sulfuric acids, (e.g., formic acid, acetic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, lactic acid, tartaric acid, malic acid, citric acid, gluconic acid, ascorbic acid, nicotinic acid, isonicotinic acid, methane-or ethanesulfonic acid, ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, naphthalenemono-and-disulfonic acids, and laurylsulfuric acid).
  • sulfonic or sulfuric acids
  • Nitrogen-containing copper chelators for example, trientine active agents such as, for example, trientine, can also be in the form of quaternary ammonium salts in which the nitrogen atom carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety.
  • nitrogen-containing copper chelators are in the form of a compound or buffered in solution and/or suspension to a near neutral pH much lower than the pH 14 of a solution of trientine itself.
  • trientine active agents include derivative trientine active agents, for example, trientine in combination with picolinic acid (2-pyridinecarboxylic acid). These derivatives include, for example, trientine picolinate and salts of trientine picolinate, for example, trientine picolinate HCl. These also include, for example, trientine di-picolinate and salts of trientine di-picolinate, for example, trientine di-picolinate HCl.
  • Picolinic acid moieties may be attached to trientine, for example one or more of the CH 2 moieties, using chemical techniques known in the art. Those in the art will be able to prepare other suitable derivatives, for example, trientine-PEG derivatives, which may be useful for particular dosage forms including oral dosage forms having increased bioavailablity.
  • trientine active agents include trientine analogue active agents.
  • analogues include cyclic and acyclic analogues according to the following formulae, for example:
  • Acyclic analogs of trientine are provided as follows based on the above Formula I for tetra-heteroatom acyclic analogues, where X1, X2, X3, and X4 are independently chosen from the atoms N, S or O such that,
  • R1, R2, R3, R4, R5, or R6 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R6 does not exist;
  • R1, R2, R3, R4, R5, and R6 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and n
  • R1, R2, R3, R4, or R5 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R4 does not exist and R1, R2, R3, R5, and R6 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and n3
  • R1, R2, R3, R5, or R6 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 allyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 allyl-S-protein.
  • R1 and R6 do not exist;
  • R2, R3, R4, and R5 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and
  • R2, R3, R4, or R5 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R3 and R6 do not exist;
  • R1, R2, R4, and R5 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and
  • R1, R2, R4, or R5 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R4 and R6 do not exist;
  • R1, R2, R3, and R5 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and
  • R1, R2, R3, or R5 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R3 and R4 do not exist;
  • R1, R2, R5 and R6 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and
  • R1, R2, R5, or R6 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R1 and R6 are joined together by a bridging group in the form of (CR13R14)n4, and X1, X2, X3, and X4 are independently chosen from the atoms N, S or O such that,
  • R2, R3, R4, and R5 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, n3, and n4 are independently chosen to be 2 or 3; and R
  • R2, R3, R4, or R5 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, R12, R13 or R14 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R5 does nor exist;
  • R2, R3 or R4 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, R12, R13 or R14 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R2 and R5 do not exist;
  • R3 and R4 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, n3, and n
  • R3, or R4 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, R12, R13 or R14 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R3 and R5 do not exist;
  • R2 and R4 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, n3, and n
  • R2, or R4 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, R12, R13 or R14 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R3, R4 and R5 do not exist;
  • R2 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, R12, R13 or R14 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 allyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • Tri-heteroatom acyclic analogues according to the above Formula II are provided where X1, X2, and X3 are independently chosen from the atoms N, S or O such that,
  • R1, R2, R3, R5 or R6 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, or R10 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R3 does not exist;
  • R1, R2, R5 or R6 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, or R10 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R3 does not exist;
  • R1, R2, R5, or R6 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, or R10 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • a second series of tri-heteroatom acyclic analogues according to the above Formula II are provided in which R1 and R6 are joined together by a bridging group in the form of (CR11R12)n3, and X1, X2, and X3 are independently chosen from the atoms N, S or O such that:
  • R2, R3, and R5 are independently chosen from H, CH3, C2-C10 straight chain or branched alkyl, C3-C10 cycloalkyl, C1-C6 alkyl C3-C10 cycloalkyl, aryl, mono, di, tri, tetra and penta substituted aryl, heteroaryl, fused aryl, C1-C6 alkyl aryl, C1-C6 alkyl mono, di, tri, tetra and penta substituted aryl, C1-C5 alkyl heteroaryl, C1-C6 alkyl fused aryl, CH 2 COOH, CH 2 SO 3 H, CH 2 PO(OH) 2 , CH 2 P(CH 3 )O(OH); n1, n2, and n3 are independently chosen to be 2 or 3; and R7, R8, R9, R10, R11, and
  • R2, R3, or R5 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R5 does not exist;
  • R2 or R3 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half-lives of the constructs.
  • functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R2 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • R7, R8, R9, R10, R11, or R12 may be functionalized in order to be attached to peptides, proteins, polyethylene glycols and other such chemical entities in order to modify the overall pharmacokinetics, deliverability and/or half lives of the constructs.
  • Examples of such functionalization include but are not limited to C1-C10 alkyl-CO-peptide, C1-C10 alkyl-CO-protein, C1-C10 alkyl-CO-PEG, C1-C10 alkyl-NH-peptide, C1-C10 alkyl-NH-protein, C1-C10 alkyl-NH—CO-PEG, C1-C10 alkyl-S-peptide, and C1-C10 alkyl-S-protein.
  • the analogues of the invention may be made using any of a variety of chemical synthesis, isolation, and purification methods known in the art.
  • aspects of the invention include controlled or other drug dose and drug dose delivery formulations and devices containing one or more copper chelators, for example, trientine or salts thereof.
  • the present invention includes, for example, doses and dosage forms for at least oral administration, transdermal delivery, topical application, suppository delivery, transmucosal delivery, injection (including subcutaneous administration, subdermal administration, intramuscular administration, depot administration, and intravenous administration (including delivery via bolus, slow intravenous injection, and intravenous drip), infusion devices (including implantable infusion devices, both active and passive), administration by inhalation or insufflation, buccal administration, sublingual administration, and ophthalmic administration.
  • Indications in which the doses, dose formulations, and routes of administration thereof will be useful include, for example, diabetic cardiomyopathy, diabetic acute coronary syndrome (e.g.; myocardial infarction—MI), diabetic hypertensive cardiomyopathy, acute coronary syndrome associated with impaired glucose tolerance (IGT), acute coronary syndrome associated with impaired fasting glucose (IFG), hypertensive cardiomyopathy associated with IGT, hypertensive cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with IGT, ischemic cardiomyopathy associated with IFG, ischemic cardiomyopathy associated with coronary heart disease (CHD), disorders of the heart muscle (cardiomyopathy or myocarditis) that include, for example, idiopathic cardiomyopathy, metabolic cardiomyopathy which includes diabetic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy, acute coronary syndrome not associated with any abnormality of glucose metabolism, hypertensive cardiomyopathy not associated with any abnormality of
  • the present invention also is directed to novel doses and dose formulations of one or more copper chelators, for example, trientine or salts thereof, useful for the pharmacological therapy of diseases in humans and other mammals as disclosed herein.
  • novel doses, formulations and devices of, for example, trientine enables effective treatment of these conditions, through novel and improved formulations of the drug suitable for administration to humans and other mammals.
  • the invention provides, for example, drug delivery formulations containing one or more copper chelators, for example, trientine or salts thereof.
  • the present invention is directed in part to novel delivery formulations of one or more copper chelators, for example, trientine to optimize bioavailability and to maintain plasma concentrations within the therapeutic range, including for extended periods, and results in increases in the time that trientine plasma concentrations of one or more copper chelators, for example, trientine or salts thereof, remain within a desired therapeutic range at the site or sites of action.
  • Controlled delivery preparations also optimize the drug concentration at the site of action and minimize periods of under and over medication, for example.
  • the invention also in part provides drug delivery formulations and devices containing one or more copper chelators, for example, one or more trientine active agents, including but not limited to, trientine, trientine dihydrochloride or other pharmaceutically acceptable salts thereof, the formulation being suitable for periodic administration, including once daily administration, to provide low dose controlled and/or low dose long-lasting in vivo release of a copper chelator for chelation of copper and excretion of chelated copper via the urine.
  • one or more copper chelators for example, one or more trientine active agents, including but not limited to, trientine, trientine dihydrochloride or other pharmaceutically acceptable salts thereof, the formulation being suitable for periodic administration, including once daily administration, to provide low dose controlled and/or low dose long-lasting in vivo release of a copper chelator for chelation of copper and excretion of chelated copper via the urine.
  • the invention also in part provides a drug delivery formulations and devices containing one or more copper chelators, for example, one or more trientine active agents, including but not limited to, trientine, trientine dihydrochloride or other pharmaceutically acceptable salts thereof, the formulation being suitable for periodic administration, including once daily administration, to provide enhanced bioavailability of a copper chelator for chelation of copper and excretion of chelated copper via the urine.
  • one or more copper chelators for example, one or more trientine active agents, including but not limited to, trientine, trientine dihydrochloride or other pharmaceutically acceptable salts thereof, the formulation being suitable for periodic administration, including once daily administration, to provide enhanced bioavailability of a copper chelator for chelation of copper and excretion of chelated copper via the urine.
  • controlled drug formulations useful for delivery of the compounds and formulations of the invention are found in, for example, Sweetman, S. C. (Ed.). Martindale. The Complete Drug Reference, 33rd Edition, Pharmaceutical Press, Chicago, 2002, 2483 pp.; Aulton, M. E. (Ed.) Pharmaceutics. The Science of Dosage Form Design. Churchill Livingstone, Edinburgh, 2000, 734 pp.; and, Ansel, H. C., Allen, L. V. and Popovich, N. G. Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott 1999, 676 pp. Excipients employed in the manufacture of drug delivery systems are described in various publications known to those skilled in the art including, for example, Kibbe, E. H.
  • the USP also provides examples of modified-release oral dosage forms, including those formulated as tablets or capsules. See, for example, The United States Pharmacopeia 23/National Formulary 18, The United States Pharmacopeial Convention, Inc., Rockville Md., 1995 (hereinafter “the USP”), which also describes specific tests to determine the drug release capabilities of extended-release and delayed-release tablets and capsules.
  • the USP test for drug release for extended-release and delayed-release articles is based on drug dissolution from the dosage unit against elapsed test time. Descriptions of various test apparatus and procedures may be found in the USP.
  • the individual monographs contain specific criteria for compliance with the test and the apparatus and test procedures to be used. Examples have been given, for example for the release of aspirin from Aspirin Extended-release Tablets (for example, see: Ansel, H. C., Allen, L. V. and Popovich, N. G. Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott 1999, p. 237). Modified-release tablets and capsules must meet the USP standard for uniformity as described for conventional dosage units. Uniformity of dosage units may be demonstrated by either of two methods, weight variation or content uniformity, as described in the USP. Further guidance concerning the analysis of extended release dosage forms has been provided by the F.D.A. (see Guidance for Industry.
  • Extended release oral dosage forms development, evaluation, and application of in vitro/in vivo correlations. Rockville, Md.: Center for Drug Evaluation and Research, Food and Drug Administration, 1997). Compliance of a dosage regime is always essential in order to derive the best benefit from a treatment regime.
  • the present invention recognizes an additional benefit from dosage forms that can provide such levels of delivery to a subject as are required to elicit the advantages now seen from the prospect of lower overall dose delivery of trientine formulations when one compares them to BID (two times a day), TID (three times a day), QID (four times a day), and so on, multiple dosage oral regimes hitherto used with, for example, trientine formulations for Wilson's disease.
  • aspects of the invention also include various drug delivery systems for the delivery of one or more copper chelators, for example, trientine or salts thereof
  • the present invention also is directed to novel types of drug delivery systems.
  • modified-release (MR) dosage forms of the present invention including delayed-release (D)R) forms; prolonged-action (PA) forms; controlled-release (CR) forms; extended-release (ER) forms; timed-release (TR) forms; and long-acting (LA) forms.
  • rate-controlled delivery is applied to certain types of drug delivery systems in which the rate of drug delivery is controlled by features of the device rather than by physiological or environmental conditions such as gastrointestinal pH or drug transit time through the gastrointestinal tract
  • formulations effect (1) delayed total drug release form some time after drug administration, (2) drug release in small aliquots intermittently after administration, (3) drug release slowly at a controlled rate governed by the delivery system, (4) drug release at a constant rate that does not vary, and/or (5) drug release for a significantly longer period than usual formulations.
  • modified”, “delayed”, “slow”, “prolonged”, “timed”, “long-acting”, “controlled”, and/or “extended” release dosage units as used herein are any appropriate delivery form.
  • compositions for administration of one or more copper chelators for example, trientine or salts thereof, include convenience to the subject; increased compliance and achievement of steady state drug levels with twice-daily (or less) dosing; smoothening of plasma drug profiles to a constant level for extended time periods; prevention of drug toxicity; and elimination of breakthrough of therapeutic failure, especially at night.
  • Modified-release dosage forms of the invention include dosage forms having drug release features based on time, course, and/or location which are designed to accomplish therapeutic or convenience objectives not offered by conventional or immediate-release forms. See, for example, Bogner, R. H. Bioavailability and bioequivalence of extended-release oral dosage forms. U.S.
  • Extended-release dosage forms of the invention include, for example, as defined by The United States Food and Drug Administration (F. D. A.), a dosage form that one that allows a reduction in dosing frequency to that presented by a conventional dosage form, e.g., a solution or an immediate-release dosage form. See, for example, Bogner, R. H. Bioavailability and bioequivalence of extended-release oral dosage forms. US Pharmacist 22 (Suppl.):3-12 (1997); Guidance for industry.
  • F. D. A. The United States Food and Drug Administration
  • Extended release oral dosage forms development, evaluation, and application of the in vitro/in vivo correlations.
  • Rockville, MD Center for Drug Evaluation and Research, Food and Drug Administration (1997).
  • Repeat action dosage forms of the invention include, for example, forms that contain two single doses of medication, one for immediate release and the second for delayed release.
  • Bi-layered tablets for example, may be prepared with one layer of drug for immediate release with the second layer deigned to release drug later as either a second dose or in an extended-release manner.
  • Targeted-release dosage forms of the invention include, for example, formulations that facilitate drug release and which are directed towards isolating or concentrating a drug in a body region, tissue, or site for absorption or for drug action.
  • dosage units for transdermal delivery of the compounds and formulations of the invention include transdermal patches, transdermal bandages, and the like.
  • dosage units for topical delivery of the compounds and formulations of the invention are any lotion, stick, spray, ointment, paste, cream, gel, etc. whether applied directly to the skin or via an intermediary such as a pad, patch or the like but which again has a slow release action in delivery of the active agent into the body of the subject.
  • dosage units for suppository delivery of the compounds and formulations of the invention include any solid dosage form inserted into a bodily orifice particularly those inserted rectally, vaginally and urethrally.
  • dosage units for transmucosal delivery of the compounds and formulations of the invention include depositories solutions for enemas, pessaries, tampons, creams, gels, pastes, foams, nebulised solutions, powders and similar formulations containing in addition to the active: ingredients such carriers as are known in the art to be appropriate.
  • dosage units for injection of the compounds and formulations of the invention include delivery via bolus such as single or multiple administrations by intravenous injection, subcutaneous, subdermal, and intramuscular administration or oral administration.
  • dosage units for depot administration of the compounds and formulations of the invention include pellets or small cylinders of active agent or solid forms wherein the active agent is entrapped in a matrix of biodegradable polymers, microemulsions, liposomes or is microencapsulated.
  • infusion devices for compounds and formulations of the invention include infusion pumps containing one or more copper chelators, for example, for example, trientine or salts thereof, at a desired amount for a desired number of doses or steady state administration, and include implantable drug pumps.
  • implantable infusion devices include any solid form in which the active agent is encapsulated within or dispersed throughout a biodegradable polymer or synthetic, polymer such as silicone, silicone rubber, silastic or similar polymer.
  • dosage units for inhalation or insufflation of the compounds and formulations of the invention include compositions comprising solutions and/or suspensions in pharmaceutically acceptable, aqueous, or organic solvents, or mixture thereof and/or powders.
  • dosage units for buccal delivery of the compounds and formulations of the invention include lozenges, tablets and the like, compositions comprising solutions and/or suspensions in pharmaceutically acceptable, aqueous, or organic solvents, or mixture thereof and/or powders
  • dosage units for sublingual delivery of the compounds and formulations of the invention include lozenges, tablets and the like, compositions comprising solutions and/or suspensions in pharmaceutically acceptable, aqueous, or organic solvents, or mixture thereof and/or powders
  • dosage units for opthalmic delivery of the compounds and formulations of the invention include compositions comprising solutions and/or suspensions in pharmaceutically acceptable, aqueous, or organic solvents, inserts,
  • the invention in part provides dose delivery devices and formulations incorporating one or more copper chelators, for example, trientine or salts thereof, complexed with one or more suitable anions to yield complexes that are only slowly soluble in body fluids.
  • one or more copper chelators for example, trientine or salts thereof
  • trientine salts of tannic acid provide for this quality, and are expected to possess utility for the treatment of conditions in which increased copper plays a role.
  • equivalent products are provided by those having the tradename Rynatan (Wallace: see, for example, Madan, P. L., “Sustained release dosage forms,” U.S. Pharmacist 15:39-50 (1990); Ryna-12 S, which contains a mixture of mepyramine tannate with phenylephrine tannate, Martindale 33rd Ed., 2080.4).
  • coated beads, granules or microspheres containing one or more copper chelators, for example, trientine or salts thereof also provides a method to achieve modified release of one or more copper chelators, for example, trientine or salts thereof, by incorporation of the drug into coated beads, granules, or microspheres.
  • Such formulations of one or more copper chelators, for example trientine or salts thereof have utility for the treatment of diseases in humans and other mammals in which a copper chelator, for example, trientine, is indicated.
  • the drug is distributed onto beads, pellets, granules or other particulate systems.
  • a solution of the drug substance is placed onto small inert nonpareil seeds or beads made of sugar and starch or onto microcrystalline cellulose spheres.
  • the nonpareil seeds are most often in the 425 to 850 micrometer range whereas the microcrystalline cellulose spheres are available ranging from 170 to 600 micrometers (see Ansel, H. C., Allen, L. V. and Popovich, N. G. Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott 1999, p. 232).
  • the microcrystalline spheres are considered more durable during production than sugar-based cores (see: Celphere microcrystalline cellulose spheres. Philadelphia: FMC Corporation, 1996).
  • Some of these granules may remain uncoated to provide immediate drug release.
  • Other granules (about two-thirds to three-quarters) receive varying coats of a lipid material such as beeswax, carnauba wax, glycerylmonostearate, cetyl alcohol, or a cellulose material such as ethylcellulose (infra).
  • a lipid material such as beeswax, carnauba wax, glycerylmonostearate, cetyl alcohol, or a cellulose material such as ethylcellulose (infra).
  • granules of different coating thickness are blended to achieve a mixture having the desired drug-release characteristics.
  • the coating material may be coloured with one or more dyes to distinguish granules or beads of different coating thickness (by depth of colour) and to provide distinctiveness to the product.
  • the granules may be placed in capsules or tableted.
  • Various coating systems are commercially available which are aqueous-based and which use ethylcellulose and plasticizer as the coating material (e.g., AquacoatTM [FMC Corporation, Philadelphia] and SurereleaseTM [Colorcon]; Aquacoat aqueous polymeric dispersion. Philadelphia: FMC Corporation, 1991; Surerelease aqueous controlled release coating system. West Point, Pa.: Colorcon, 1990; Butler, J., Cumming, I, Brown, J. et al. A novel multiunit controlled-release system. Pharm Tech 22:122-138 (1998); Yazici, E., Oner, L., Kas, H. S. & Hincal, A. A. Phenytoin sodium microspheres: bench scale formulation, process characterization and release kinetics.
  • AquacoatTM FMC Corporation, Philadelphia
  • SurereleaseTM Colorcon
  • Aqueous-based coating systems eliminate the hazards and environmental concerns associated with organic solvent-based systems. Aqueous and organic solvent-based coating methods have been compared (see, for example, Hogan, J. E. Aqueous versus organic solvent coating. Int J Pharm Tech Prod Manufacture 3:17-20 (1982)).
  • the variation in the thickness of the coats and in the type of coating materials used affects the rate at which the body fluids are capable of penetrating the coating to dissolve the drug. Generally, the thicker the coat, the more resistant to penetration and the more delayed will be drug release and dissolution.
  • the coated beads are about 1 mm in diameter.
  • Each of Eudragit® RS and Eudragit® RL is an ammonio methacrylate copolymer.
  • the release rate can be controlled not only by incorporating therein suitable water-soluble pore formers, such as lactose, mannitol, sorbitol, etc., but also by the thickness of the coating layer applied.
  • Multi tablets include small spheroid-shaped compressed minitablets that may have a diameter of between 3 to 4 mm and can be placed in gelatin capsule shell to provide the desired pattern of drug release.
  • Each capsule may contain 8-10 minitablets, some uncoated for immediate release and others coated for extended drug release.
  • the following methods may be employed to generate delivery systems containing modified-release delivery forms of one or more copper chelators, for example trientine or salts thereof or other trientine active agents, suitable for oral administration to humans and other mammals.
  • Two basic mechanisms are available to achieve modified release drug delivery. These are altered dissolution or diffusion of drugs and excipients.
  • four processes may be employed, either simultaneously or consecutively. These are as follows: (i) hydration of the device (e.g., swelling of the matrix); (ii) diffusion of water into the device; (iii) controlled or delayed dissolution of the drug; and (iv) controlled or delayed diffusion of dissolved or solubilized drug out of the device.
  • Modified release dosage forms commonly fit into one of three categories of system,: monolithic or matrix; reservoir- or membrane-controlled; or osmotic pump systems. Each comprises the following components: active drug; release controlling agents; matrix modifiers; drug modifiers; supplementary coatings; and conventional formulation excipients, such as those described in reference works known to those skilled in the art (see, for example, Kibble A. H (ed.) Handbook of Pharmaceutical Excipients, 3rd Edition, American Pharmaceutical Association, 2000, 665 pp.).
  • extended drug action may be achieved by affecting the rate at which the drug is released from the dosage form and/or by slowing the transit time of the dosage form through the gastrointestinal tract (see Bogner, R. H. Bioavailability and bioequivalence of extended-release oral dosage forms. US Pharmacist 22 (Suppl.):3-12 (1997)).
  • the rate of drug release from solid dosage forms may be modified by the technologies described below which, in general, are based on the following: 1) modifying drug dissolution by controlling access of biologic fluids to the drug through the use of barrier coatings; 2) controlling drug diffusion rates from dosage forms; and 3) chemically reacting or interacting between the drug substance or its pharmaceutical barrier and site-specific biological fluids.
  • the active agent is either coated or entrapped in a substance that is slow digested or dispersed into the intestinal tract.
  • the rate of availability of the active agent is a function of the rate of digestion of the dispersible material. Therefore, the release rate, and thus the effectiveness of the agent, varies from subject to subject depending upon the ability of the subject to digest the material.
  • the active agent is dispersed in a water-soluble colloid and then coated with a rupturable plastic, non-digestible material that is permeable to the diffusion of water.
  • U.S. Pat. No. 3,115,441 discloses another encapsulation method useful for delivery of the compounds and formulations of the invention wherein particles of active agent are first given a quick thin coating of a film-forming material and a non-toxic, hydrophobic material that is then coated with successive coatings of an organic solvent-resistant material. The coated particles are mixed with uncoated active agent and this mixture is then formed into a tablet with the coated tablets being entrapped in a matrix of the uncoated active agent. Tablets made according to this method have the advantage of providing immediate delivery of the compounds and formulations of the invention because the matrix material (which comprises the initial dosage) dissolves immediately upon ingestion.
  • Another approach is to provide an improved blood level profile of the compounds and formulations of the invention that results from simply applying a film of a non-aqueous solution of cellulose acetate over either individual particles of active agent before tableting or over the outside of tablets formed from untreated active agent particles, which upon drying forms a coating of cellulose acetate.
  • the film-forming agent may be supplemented with: plasticizers (such as polyoxyethylene glycols of high molecular weight, esters of polyacids such as citric acid or phthalic acid) fillers (such as talc, metal oxides such as titanium oxide) colorants chosen from those usable and approved by the pharmaceutical and food industries.
  • plasticizers such as polyoxyethylene glycols of high molecular weight, esters of polyacids such as citric acid or phthalic acid
  • fillers such as talc, metal oxides such as titanium oxide
  • a further form of slow release form of the compounds and formulations of the invention is any suitable osmotic system where semipermeable membranes of cellulose acetate, cellulose acetate butyrate, cellulose acetate propionate, to control the release of active ingredients. These can be coated with aqueous dispersions of enteric lacquers without changing release rate.
  • An example of such an osmotic system is an osmotic pump device, an example of which is the OrosTM device developed by Alza Inc. (U.S.A.). This system comprises a core tablet surrounded by a semi-permeable membrane coating having a 0.4 mm diameter hole produced by a laser beam.
  • the core tablet has two layers, one containing the drug (the “active” layer) and the other containing a polymeric osmotic agent (the “push” layer).
  • the core layer consists of active drug, filler, a viscosity modulator, and a solubilizer.
  • the system operates on the principle of osmotic pressure. This system is suitable for delivery of a wide range of drugs, including trientine or salts thereof.
  • the coating technology is straightforward, and release is zero-order.
  • the semi-permeable membrane permits aqueous fluid to enter from the stomach into the core tablet, dissolving or suspending the drug.
  • the osmotic layer As pressure increases in the osmotic layer, it forces or pumps the drug solution out of the delivery orifice on the side of the tablet Only the drug solution (not the undissolved drug) is capable of passing through the hole in the tablet.
  • the system is designed such that only a few drops of water are drawn into the tablet each hour.
  • the rate of inflow of aqueous fluid and the function of the tablet depends on the existence of an osmotic gradient between the contents of the bi-layer and the fluid in the gastrointestinal tract.
  • Drug delivery is essentially constant as long as the osmotic gradient remains unchanged.
  • the drug release rate may be altered by changing the surface area, the thickness or composition of the membrane, and/or by changing the diameter of the drug release orifice.
  • the drug-release rate is not affected by gastrointestinal acidity, alkalinity, fed conditions, or gut motility.
  • the biologically inert components of the tablet remain intact during gut transit and are eliminated in the feces as an insoluble shell.
  • Other examples of the application of this technology are provided by Glucotrol XL Extended Release Tablets (Pfizer Inc.) and Procardia XL Extended Release Tablets (Pfizer Inc.; see, Martindale 33rd Ed., p. 2051.3).
  • the invention also provides delivery devices for compounds and formulations of the invention that utilize monolithic matrices including, for example, slowly eroding or hydrophilic polymer matrices, in which one or more copper chelators, for example, trientine or salts thereof, is compressed or embedded.
  • monolithic matrices including, for example, slowly eroding or hydrophilic polymer matrices, in which one or more copper chelators, for example, trientine or salts thereof, is compressed or embedded.
  • Monolithic matrix devices for delivery of the compounds and formulations of the invention comprise those formed using either of the following systems, for example: (I), drug particles are dispersed in a soluble matrix, in which they become increasingly available as the matrix dissolves or swells; examples include hydrophilic colloid matrices, such as hydroxypropylcellulose (BP) or hydroxypropyl cellulose (USP); hydroxypropyl methylcellulose (HPMC; BP, USP); methylcellulose (MC; BP, USP); calcium carboxymethylcellulose (Calcium CMC; BP, USP); acrylic acid polymer or carboxy polymethylene (Carbopol) or Carbomer (BP, USP); or linear glycuronan polymers such as alginic acid (BP, USP), for example those formulated into microparticles from alginic acid (alginate)-gelatin hydrocolloid coacervate systems, or those in which liposomes have been encapsulated by coatings of alginic acid with poly
  • Drug release occurs as the polymer swells, forming a matrix layer that controls the diffusion of aqueous fluid into the core and thus the rate of diffusion of drug from the system.
  • the rate of drug release depends upon the tortuous nature of the channels within the gel, and the viscosity of the entrapped fluid, such that different release kinetics can be achieved, for example, zero-order, or first-order combined with pulsatile release.
  • gels are not cross-linked, there is a weaker, non-permanent association between the polymer chains, which relies on secondary bonding. With such devices, high loading of the active drug is achievable, and effective blending is frequent.
  • Hydrophilic matrix devices typically contain pH buffers, surfactants, counter-ions, lubricants such as magnesium stearate (BP, USP) and a glidant such as colloidal silicon dioxide (USP; colloidal anhydrous silica, BP) in addition to drug substance and hydrophilic matrix; (II) drug particles are dissolved in an insoluble matrix, from which drug becomes available as solvent enters the matrix, often through channels, and dissolves the drug particles.
  • Examples include systems formed with a lipid matrix, or insoluble polymer matrix, including preparations formed from Carnauba wax (BP; USP); medium-chain triglyceride such as fractionated cocoanut oil (BP) or triglycerida saturata media (PhEur); or cellulose ethyl ether or ethylcellulose (BP, USP).
  • BP Carnauba wax
  • medium-chain triglyceride such as fractionated cocoanut oil (BP) or triglycerida saturata media (PhEur); or cellulose ethyl ether or ethylcellulose (BP, USP).
  • Lipid matrices are simple and easy to manufacture, and incorporate the following blend of powdered components: lipids (20-40% hydrophobic solids w/w) which remain intact during the release process; drug substance; channeling agent, such as sodium chloride or sugars, which leaches from the formulation, forming aqueous micro-channels (capillaries) through which solvent enters, and through which drug is released.
  • channeling agent such as sodium chloride or sugars
  • the drug is embedded in an inert insoluble polymer and is released by leaching of aqueous fluid, which diffuses into the core of the device through capillaries formed between particles, and from which drug diffuses out of the device.
  • the rate of release is controlled by the degree of compression, particle size, and the nature and relative content (w/w) of excipients.
  • An example of such a device is that of Ferrous Gradumet (Martindale 33rd Ed., 1360.3).
  • a further example of a suitable insoluble matrix is an inert plastic matrix.
  • trientine active agent is granulated with an inert plastic material such as polyethylene, polyvinyl acetate, or polymethacrylate, and the granulated mixture is then compressed into tablets. Once ingested, the drug is slowly released from the inert plastic matrix by diffusion (see, for example, Bodmeier, R.
  • An immediate-release portion of drug may be compressed onto the surface of the tablet.
  • the inert tablet matrix, expended of drug, is excreted with the feces.
  • An example of a successful dosage form of this type is Gradumet (Abbott; see, for example, Ferro-Gradumet, Martindale 33rd Ed., p. 1860.4).
  • dosage forms of the compounds and formulations of the invention in which the drug is bound to a biocompatible polymer by a labile chemical bond, e.g., polyanhydrides prepared from a substituted anhydride (itself prepared by reacting an acid chloride with the drug: methacryloyl chloride and the sodium salt of methoxy benzoic acid) have been used to form a matrix with a second polymer (Eudragit RL) which releases drug on hydrolysis in gastric fluid (see: Chafi, N., Montheard, J. P. & Vergnaud, J. M. Release of 2-aminothiazole from polymeric carriers. Int J Pharm 67:265-274 (1992)).
  • a labile chemical bond e.g., polyanhydrides prepared from a substituted anhydride (itself prepared by reacting an acid chloride with the drug: methacryloyl chloride and the sodium salt of methoxy benzoic acid) have been used to form a matrix with a second
  • the polymer selected for use must form a gelatinous layer rapidly enough to protect the inner core of the tablet from disintegrating too rapidly after ingestion.
  • the proportion of polymer is increased in a formulation so is the viscosity of the gel formed with a resulting decrease in the rate of drug diffusion and release (see Formulating for controlled release with Methocel Premium cellulose ethers. Midland, Mich.: Dow Chemical Company, 1995).
  • 20% (w/w) of HPMC results in satisfactory rates of drug release for an extended-release tablet formulation.
  • consideration must be given to the possible effects of other formulation ingredients such as fillers, tablet binders, and disintegrants.
  • An example of a proprietary product formulated using a hydrophilic matrix base of HPMC for extended drug release is that of Oramorph SR Tablets (Roxane; see Martindale 33rd Ed., p. 2014.4).
  • Two-layered tablets can be manufactured containing one or more of the compounds and formulations of the invention, with one layer containing the uncombined drug for immediate release and the other layer having the drug imbedded in a hydrophilic matrix for extended-release.
  • Three-layered tablets may also be similarly prepared, with both outer layers containing the drug for immediate release.
  • Some commercial tablets are prepared with an inner core containing the extended-release portion of drug and an outer shell enclosing the core and containing drug for immediate release.
  • the invention also provides forming a complex between the active agent, e.g., one or more compounds and formulations of the invention and an ion exchange resin, whereupon the complex may be tableted, encapsulated or suspended in an aqueous vehicle. Release of the active agent is dependent on the local pH and electrolyte concentration such that the choice of ion exchange resin may be made so as to preferentially release the active agent in a given region of the alimentary canal. Delivery devices incorporating such a complex are also provided. For example, a modified release dosage form of trientine can be produced by the incorporation of trientine into complexes with an anion-exchange resin.
  • Solutions of trientine may be passed through columns containing an ion-exchange resin to form a complex by the replacement of H 3 O + ions.
  • the resin-trientine complex is then washed and may be tableted, encapsulated, or suspended in an aqueous vehicle.
  • the release of the trientine is dependent on the pH and the electrolyte concentration in the gastrointestinal fluid. Release is greater in the acidity of the stomach than in the less acidic environment of the small intestine.
  • Alternative examples of this type of extended release preparation are provided by hydrocodone polistirex and chorpheniramine polistirex suspension (Medeva; Tussionex Pennkinetic Extended Release Suspension, see: Martindale 33rd Ed., p.
  • Such resin-trientine active agent systems can additionally incorporate polymer barrier coating and bead technologies in addition to the ion-exchange mechanism.
  • the initial dose comes from an uncoated portion, and the remainder from the coated beads.
  • the coating does not dissolve, and release may be extended over a 12-hour period by ion exchange.
  • the drug containing particles are minute, and may be also suspended to produce a liquid with extended-release characteristics, as well as solid dosage forms.
  • Such preparations may also be suitable for administration, for example in depot preparations suitable for intramuscular injection.
  • the invention also provides a method to produce modified release preparations of one or more copper chelators, for example, trientine or salts thereof, by microencapsulation.
  • microencapsulated preparations are useful for the treatment of humans and other mammals, in which copper chelation therapy is indicated.
  • Microencapsulation is a process by which solids, liquids, or even gasses may be encapsulated into microscopic size particles through the formation of thin coatings of “wall” material around the substance being encapsulated such as disclosed in U.S. Pat. Nos. 3,488,418; 3,391,416 and 3,155,590.
  • Gelatin is commonly employed as a wall-forming material in microencapsulated preparations, but synthetic polymers such as polyvinyl alcohol (USP), ethylcellulose (BP, USP), polyvinyl chloride, and other materials may also be used (see, for example, Zentner, G. M., Rork, G. S. & Himmelstein, K. J. Osmotic flow through controlled porosity films: an approach to delivery of water soluble compounds. J Controlled Release 2:217-229 (1985); Fites, A. L., Banker, G. S. & Smolen, V. F. Controlled drug release through polymeric films.
  • synthetic polymers such as polyvinyl alcohol (USP), ethylcellulose (BP, USP), polyvinyl chloride, and other materials may also be used (see, for example, Zentner, G. M., Rork, G. S. & Himmelstein, K. J. Osmotic flow through controlled porosity films: an approach to delivery of water
  • Encapsulation begins with the dissolving of the prospective wall material, say gelatin, in water.
  • One or more copper chelators for example, trientine or one or more salts thereof, is then added and the two-phase mixture is thoroughly stirred.
  • a solution of a second material is added, that can be acacia (BP, USP). This additive material is chosen to have the ability to concentrate the gelatin (polymer) into tiny liquid droplets.
  • Different rates of trientine release may be obtained by changing the core-to-wall ratio, the polymer used for the coating, or the method of microencapsulation (for example, see: Yazici, E., Oner, L., Kas, H. S. & Hincal, A. A. Phenytoin sodium microspheres: bench scale formulation, process characterization and release kinetics. Pharmaceut Dev Technol 1996;1:175-183).
  • microencapsulation the administered dose of one or more copper chelators, for example, trientine or salts thereof, is subdivided into small units that are spread over a large area of the gastrointestinal tract, which may enhance absorption by diminishing localized drug concentrations (see Yazici et al., supra).
  • An example of a drug that is commercially available in a microencapsulated extended-release dosage form is potassium chloride (Micro-K Exten-caps, Wyeth-Ayerst, Martindale 33rd Ed., p 1968.1).
  • the invention also includes repeat action tablets containing one or more copper chelators, for example, trientine or salts thereof.
  • Further examples of a method by which modified release forms of one or more copper chelators, for example, trientine or salts thereof, suitable for treatment of humans or other mammals, can be produced are provided by the incorporation of trientine into repeat action tablets. These are prepared so that an initial dose of the drug is released immediately followed later by a second dose.
  • the tablets may be prepared with the immediate-release dose in the tablet's outer shell or coating with the second dose in the tablet's inner core, separated by a slowly permeable barrier coating. In general, the drug from the inner core is exposed to body fluids and released 4 to 6 hours after administration.
  • Repeat action dosage forms are suitable for the administration of one or more copper chelators, for example, trientine or salts thereof, for the indications noted herein, including but not limited to chronic conditions such as heart failure, diabetic heart disease, acute coronary syndrome, hypertensive heart disease, ischemic heart disease, coronary artery disease, peripheral arterial disease, or any form of cancer.
  • This form of delivery is particularly suitable for delivery of trientine, since it has a rapid rate of absorption and excretion.
  • the invention also includes delayed-release oral dosage forms containing one or more copper chelators, for example, trientine or salts thereof.
  • the release of one or more copper chelators, for example, trientine or salts thereof from an oral dosage form can be intentionally delayed until it reaches the intestine by way of, for example, enteric coating.
  • Enteric coatings by themselves are not an efficient method for the delivery of copper chelators such as, for example, trientine or salts thereof including trientine dihydrochloride, because of the inability of such coating systems to provide or achieve a sustained therapeutic effect after release onset.
  • Enteric coats are designed to dissolve or breakdown in an alkaline environment. The presence of food may increase the pH of the stomach.
  • enteric-coated trientine dihydrochloride with food or the presence of food in the stomach may lead to dose dumping and unwanted secondary effects.
  • trientine dihydrochloride can give rise to gastrointestinal side-effects, it would be desirable to have a drug delivery system that is capable of providing the controlled delivery of trientine dihydrochloride or other pharmaceutically acceptable salts of trientine in a predictable manner over a long period of time.
  • Enteric coatings also have application in the present invention when combined or incorporated with one or more of the other dose delivery formulations or devices described herein.
  • This form of delivery conveys the advantage of minimizing the gastric irritation that may be caused in some subjects by trientine.
  • the enteric coating may be time-dependent, pH-dependent where it breaks down in the less acidic environment of the intestine and erodes by moisture over time during gastrointestinal transit, or enzyme-dependent where it deteriorates due to the hydrolysis-catalyzing action of intestinal enzymes (see, for example, Bengal, N. A., et al. “Modifying the release properties of Eudragit L30D,” Drug Dev Ind Pharm. 17:2497-2509 (1991)).
  • enteric coat tablets and capsules are fats including triglycerides, fatty acids, waxes, shellac, and cellulose acetate phthalate although further examples of enteric coated preparations can be found in the USP.
  • the invention also provides drug delivery devices incorporating one or more copper chelators, for example, trientine or salts thereof, in a membrane-control system.
  • Such devices comprise a rate-controlling membrane surrounding a drug reservoir. Following oral administration the membrane gradually becomes permeable to aqueous fluids, but does not erode or swell.
  • the drug reservoir may be composed of a conventional tablet, or a microparticle pellet containing multiple units that do not swell following contact with aqueous fluids.
  • the cores dissolve without modifying their internal osmotic pressure, thereby avoiding the risk of membrane rupture, and typically comprise 60:40 mixtures of lactulose: microcrystalline cellulose (w/w).
  • Multiple-unit membrane-controlled systems typically comprise more than one discrete unit. They can contain discrete spherical beads individually coated with rate-controlling membrane and may be encapsulated in a hard gelatin shell (examples of such preparations include Contac 400; martindale 33rd Ed., 1790.1 and Feospan; Martindale 33rd Ed., p. 1859.4). Alternatively, multiple-unit membrane-controlled systems may be compressed into a tablet (for example, Suscard; Martindale 33rd Ed., p. 2115.1).
  • Alternative implementations of this technology include devices in which the drug substance is coated around inert sugar spheres, and devices prepared by extrusion spheronization employing a conventional matrix system. Advantages of such systems include the more consistent gastrointestinal transit rate achieved by multiple-unit systems, and the fact that such systems infrequently suffer from catastrophic dose dumping. They are also ideal for the delivery of more than one drug at a time.
  • Preferred for oral delivery is a sustained release form of one or more compounds and formulations of the invention which is a matrix formation, such a matrix formation taking the form of film coated spheroids containing as active ingredient one or more copper chelators, for example, trientine or salts thereof such as trientine dihydrochloride, and a non water soluble spheronising agent.
  • spheroid is known in the pharmaceutical art and means spherical granules having a diameter usually of between 0.01 mm and 4 mm.
  • the spheronising agent may be any pharmaceutically acceptable material that, together with the active ingredient, can be spheronised to form spheroids. Microcrystalline cellulose is preferred.
  • Suitable microcrystalline cellulose includes, for example, the material sold as Avicel PH 101 (Trade Mark, FMC Corporation).
  • the film-coated spheroids contain between 70% and 99% (by wt), especially between 80% and 95% (by wt), of the spheronising agent, especially microcrystalline cellulose.
  • the spheroids may also contain a binder. Suitable binders, such as low viscosity, water soluable polymers, will be well known to those skilled in the pharmaceutical art.
  • a suitable binder is, in particular polyvinylpyrrolidone in various degrees of polymerization.
  • the spheroids may contain a water insoluble polymer, especially an acrylic polymer, an acrylic copolymer, such as a methacrylic acid-ethyl acrylate copolymer, or ethyl cellulose.
  • thickening agents or binders include:the lipid type, among which are vegetable oils (cotton seed, sesame and groundnut oils) and derivatives of these oils (hydrogenated oils such as hydrogenated castor oil, glycerol behenate,the waxy type such as natural carnauba wax or natural beeswax, synthetic waxes such as cetyl ester waxes, the amphiphilic type such as polymers of ethylene oxide (polyoxyethylene glycol of high molecular weight between 4000 and 100000) or propylene and ethylene oxide copolymers (poloxamers), the cellulosic type (semisynthetic derivatives of cellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, hydroxymethylcellulose, of high molecular weight and high viscosity, gum) or any other polysaccharide such as alginic acid, the polymeric type such as acrylic acid polymers (such as carbomers), and the mineral type such as colloidal silica, bentonite.
  • Suitable diluents for the active ingredient in the pellets, spheroids or core are, e.g., microcrystalline cellulose, lactose, dicalcium phosphate, calcium carbonate, calcium sulphate, sucrose, dextrates, dextrin, dextrose, dicalcium phosphate dihydrate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, cellulose, microcrystalline cellulose, sorbitol, starches, pregelatinized starch, talc, tricalcium phosphate and lactose.
  • Suitable lubricants are e.g., magnesium stearate and sodium stearyl fumarate.
  • Suitable binding agents are e.g., hydroxypropyl methyl cellulose, polyvidone and methyl cellulose.
  • Suitable binders that may be included are: gum arabic, gum tragacanth, guar gum, alginic acid, sodium alginate, sodium carboxymethylcellulose, dextrin, gelatin, hydroxyethylcellulose, hydroxypropylcellulose, liquid glucose, magnesium and aluminium.
  • Suitable disintegrating agents are starch, sodium starch glycolate, crospovidone and croscarmalose sodium.
  • Suitable surface active are Poloxamer 188®, polysorbate 80 and sodium lauryl sulfate.
  • Suitable flow aids are talc colloidal anhydrous silica.
  • Suitable lubricants that may be used are glidants (such as anhydrous silicate, magnesium trisilicate, magnesium silicate, cellulose, starch, talc or tricalcium phosphate) or alternatively antifriction agents (such as calcium stearate, hydrogenated vegetable oils, paraffin, magnesium stearate, polyethylene glycol, sodium benzoate, sodium lauryl sulphate, fumaric acid, stearic acid or zinc stearate and talc).
  • Suitable water-soluble polymers are PEG with molecular weights in the range 1000 to 6000.
  • lubricants and nonstick agents which may be mentioned, are higher fatty acids and their alkali metal and alkaline-earth-metal salts, such as calcium stearate.
  • Suitable disintegrants are, in particular, chemically inert agents.
  • Disintegrants that may be mentioned as preferred are cross-linked polyvinylpyrrolidone, cross-linked sodium carboxymethylcelluloses, and sodium starch glycolate.
  • the dosage unit if oral preferably delivers more than about than 50% of a copper chelator, for example, trientine dihydrochloride, in 12 hrs at a pH of about ⁇ 6.5 in a controlled manner during in vivo and in vitro dissolution.
  • a copper chelator for example, trientine dihydrochloride
  • Still further embodiments of the invention include forms of one or more copper chelators, for example, trientine or salts thereof, incorporated into transdermal drug delivery systems, such as those described in: Transdermal Drug Delivery Systems, Chapter 10. In: Ansel, H. C., Allen, L. V. and Popovich, N. G. Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott 1999, pp. 263-278). Transdermal drug delivery systems facilitate the passage of therapeutic quantities of drug substances through the skin and into the systemic circulation to exert systemic effects, as originally described (see Stoughton, R. D. Percutaneous absorption. Toxicol Appl Pharmacol 7:1-8 (1965)).
  • Methods known to enhance the delivery of drugs by the percutaneous route include chemical skin penetration enhancers, which increase skin permeability by reversibly damaging or otherwise altering the physicochemical nature of the stratum corneum to decrease its resistance to drug diffusion (see Shah, V. P., Peck, C. C. & Williams, R. L. Skin penetration enhancement: clinical pharmacological and regulatory considerations. In: Walters, K. A. & Hadgraft, J. (Eds.) Pharmaceutical skin penetration enhancement. New York: Dekker, 1993).
  • effective alterations are increased hydration of the stratum corneum and/or a change in the structure of the lipids and lipoproteins in the intercellular channels brought about through solvent action or denaturation (see Walters K.
  • Skin penetration enhances suitable for formulation with trientine in Transdermal Drug Delivery Systems may be chosen from the following list: acetone, laurocapram, dimethylacetamide, dimethylformamide, dimethylsulphoxide, ethanol, oleic acid, polyethylene glycol, propylene glycol and sodium lauryl sulphate. Further skin penetration enhancers may be found in publications known to those skilled in the art (see, for example, Osborne, D. W., & Henke, J. J., “Skin penetration enhancers cited in the technical literature,” Pharm Tech 21:50-66 (1997); Rolf, D., “Chemical and physical methods of enhancing transdermal drug delivery,” Pharm Tech 12:130-139 (1988)).
  • Iontophoresis involves the delivery of charged chemical compounds across the skin membrane using an applied electrical field. Such methods have proven suitable for delivery of a number of drugs.
  • another embodiment of the invention comprises one or more copper chelators, for example, trientine or salts thereof, formulated in such a manner suitable for administration by iontophoresisor sonophoresis.
  • Formulations of one or more copper chelators, for example, trientine, suitable for administration by iontophoresis or sonophoresis may be in the form of gels, creams, or lotions.
  • Transdermal delivery may utilize, among others, monolithic delivery systems, drug-impregnated adhesive delivery systems (e.g., the LatitudeTM drug-in-adhesive system from 3M), active transport devices and membrane-controlled systems.
  • Monolithic systems incorporate an active agent matrix, comprising a polymeric material in which the active agent is dispersed between backing and frontal layers.
  • Drug impregnated adhesive delivery systems comprise an adhesive polymer in which one or more compounds and formulations of the invention and any excipients are incorporated into the adhesive polymer.
  • Active transport devices incorporate an active agent reservoir, often in liquid or gel form, a membrane that may be rate controlling, and a driving force to propel the active agent across the membrane.
  • Transdermal delivery dosage forms include those which substitute the trientine active ingredient, preferably trientine dihydrochloride for the diclofenic or other pharmaceutically acceptable salt thereof referred to in the transdermal delivery systems disclosed in, by way of example, U.S. Pat. Nos. 6,193,996, 6,262,121.
  • Topical administration of one or more compounds and formulations of the invention ingredient can be prepared as an admixture or other pharmaceutical formulation to be applied in a wide variety of ways including, but are not limited to, lotions, creams gels, sticks, sprays, ointments and pastes. These product types may comprise several types of formulations including, but not limited to solutions, emulsions, gels, solids, and liposomes. If the topical composition is formulated as an aerosol and applied to the skin as a spray-on, a propellant may be added to a solution composition. Suitable propellants as used in the art can be utilized.
  • topical administration of an active agent reference is made to U.S. Pat. Nos. 5,602,125, 6,426,362 and 6,420,411.
  • sustained dosage forms in accordance with the present invention are any variants of the oral forms that are adapted for suppository or other parenteral use.
  • these compositions may be prepared by mixing one or more compounds and formulations of the invention with a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquidity and/or dissolve in the rectal cavity to release the drug.
  • Suppositories are generally solid dosage forms intended for insertion into body orifices including rectal, vaginal and occasionally urethrally and can be long acting or slow release.
  • Suppositories include a base that can include, but is not limited to, materials such as alginic acid, which will prolong the release of the pharmaceutically acceptable active ingredient over several hours (5-7).
  • bases can be characterized into two main categories and a third miscellaneous group: 1) fatty or oleaginous bases, 2) water-soluble or water-miscible bases and 3) miscellaneous bases, generally combinations of lipophilic and hydrophilic substances.
  • Fatty or oleaginous bases include hydrogenated fatty acids of vegetable oils such as palm kernel oil and cottonseed oil, fat-based compound containing compounds of glycerin with the higher molecular weight fatty acids such as palmitic and stearic acids, cocoa butter is also used where phenol and chloral hydrate lower the melting point of cocoa butter when incorporated, solidifying agents like cetyl esters wax (about 20%) or beeswax (about 4%) may be added to maintain a solid suppository.
  • Other bases include other commercial products such as Fattibase (triglycerides from palm, palm kernel and coconut oils with self-emulsifying glycerol monostearate and poloxyl stearate), Wecobee and Witepsol bases.
  • Water-soluble bases are generally glycerinated gelatin and Water-miscible bases are generally polyethylene glycols.
  • the miscellaneous bases include mixtures of the oleaginous and water-soluble or water-miscible materials.
  • An example of such a base in this group is polyoxyl 40 stearate and polyoxyethylene diols and the free glycols.
  • Transmucosal delivery of the compounds and formulations of the invention may utilize any mucosal membrane but commonly utilizes the nasal, buccal, vaginal and rectal tissues.
  • Formulations suitable for nasal administration of the compounds and formulations of the invention may be administered in a liquid form, for example, nasal spray, nasal drops, or by aerosol administration by nebulizer, including aqueous or oily solutions of the active ingredient.
  • Formulations for nasal administration, wherein the carrier is a solid include a coarse powder having a particle size, for example, of less than about 100 microns, preferably less than about 50 microns, which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • compositions in solution may be neubulised by the use of inner gases and such nebulised solutions may be breathed directly from the neulising device or the nebulising device may be attached to a facemask, tent or intermittent positive pressure-breathing machine.
  • Solutions, suspensions or powder compositions may be administered orally or nasally from devices that deliver the formulation in an appropriate manner.
  • Formulations may be prepared as aqueous solutions for example in saline, solutions employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bio-availability, fluorocarbons, and/or other solubilising or dispersing agents known in the art.
  • the invention provides extended-release formulations containing one or more copper chelators, for example, trientine or salts thereof suitable for parenteral administration.
  • Extended rates of drug action following injection may be achieved in a number of ways, including the following: crystal or amorphous drug forms having prolonged dissolution characteristics; slowly dissolving chemical complexes of the drug entity; solutions or suspensions of drug in slowly absorbed carriers or vehicles (as oleaginous); increased particle size of drug in suspension; or, by injection of slowly eroding microspheres of drug (for example, see: Friess, W., Lee, G. and Groves, M. J. Insoluble collagen matrices for prolonged delivery of proteins. Pharmaceut Dev Technol 1:185-193 (1996)).
  • the duration of action of the various forms of insulin for example is based in part on its physical form (amorphous or crystalline), complex formation with added agents, and its dosage form (solution of suspension).
  • the copper chelator must be formulated into a stable, safe pharmaceutical composition for administration to a patient.
  • the copper chelator is a trientine active agent.
  • the composition can be prepared according to conventional methods by dissolving or suspending an amount of the trientine active agent ingredient in a diluent. The amount is from between 0.1 mg to 1000 mg per ml of diluent of the trientine active agent.
  • An acetate, phosphate, citrate or glutamate buffer may be added allowing a pH of the final composition to be from 5.0 to 9.5; optionally a carbohydrate or polyhydric alcohol tonicifier and, a preservative selected from the group consisting of m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol may also be added.
  • a sufficient amount of water for injection is used to obtain the desired concentration of solution.
  • Additional tonicifying agents such as sodium chloride, as well as other excipients, may also be present, if desired. Such excipients, however, must maintain the overall tonicity of the trientine active agent.
  • buffer when used with reference to hydrogen-ion concentration or pH, refer to the ability of a system, particularly an aqueous solution, to resist a change of pH on adding acid or alkali, or on dilution with a solvent
  • Characteristic of buffered solutions which undergo small changes of pH on addition of acid or base, is the presence either of a weak acid and a salt of the weak acid, or a weak base and a salt of the weak base.
  • An example of the former system is acetic acid and sodium acetate.
  • the change of pH is slight as long as the amount of hydroxyl ion added does not exceed the capacity of the buffer system to neutralize it.
  • the stability of the parenteral formulation of the present invention is enhanced by maintaining the pH of the formulation in the range of approximately 5.0 to 9.5.
  • Other pH ranges for example, include, 5.5 to 9.0, or 6.0 to 8.5, or 6.5 to 8.0, or 7.0 to 7.5.
  • the buffer used in the practice of the present invention is selected from any of the following, for example, an acetate buffer, a phosphate buffer or glutamate buffer, the most preferred buffer being a phosphate buffer.
  • Carriers or excipients can also be used to facilitate administration of the compound.
  • carriers and excipients include calcium carbonate, calcium phosphate, various sugars such as lactose, glucose, or sucrose, or types of starch, cellulose derivatives, gelatin, polyethylene glycols and physiologically compatible solvents.
  • a stabilizer may be included in the present formulation but, and importantly, is not needed. If included, however, a stabilizer useful in the practice of the present invention is a carbohydrate or a polyhydric alcohol.
  • the polyhydric alcohols include such compounds as sorbitol, mannitol, glycerol, and polyethylene glycols (PEGs).
  • the carbohydrates include, for example, mannose, ribose, trehalose, maltose, inositol, lactose, galactose, arabinose, or lactose.
  • Suitable stabilizers include, for example, polyhydric alcohols such as sorbitol, mannitol, inositol, glycerol, xylitol, and polypropylene/ethylene glycol copolymer, as well as various polyethylene glycols (PEG) of molecular weight 200, 400, 1450, 3350, 4000, 6000, and 8000).
  • polyhydric alcohols such as sorbitol, mannitol, inositol, glycerol, xylitol, and polypropylene/ethylene glycol copolymer
  • PEG polyethylene glycols
  • USP United States Pharmacopeia
  • anti-microbial agents in bacteriostatic or fungistatic concentrations must be added to preparations contained in multiple dose containers. They must be present in adequate concentration at the time of use to prevent the multiplication of microorganisms inadvertently introduced into the preparation while withdrawing a portion of the contents with a hypodermic needle and syringe, or using other invasive means for delivery, such as pen injectors.
  • Antimicrobial agents should be evaluated to ensure compatibility with all other components of the formula, and their activity should be evaluated in the total formula to ensure that a particular agent that is effective in one formulation is not ineffective in another. It is not uncommon to find that a particular agent will be effective in one formulation but not effective in another formulation.
  • a preservative is, in the common pharmaceutical sense, a substance that prevents or inhibits microbial growth and may be added to a pharmaceutical formulation for this purpose to avoid consequent spoilage of the formulation by microorganisms. While the amount of the preservative is not great, it may nevertheless effect the overall stability of the trientine active agent. Thus, even selection of a preservative can be difficult.
  • the preservative for use in the practice of the present invention can range from 0.005 to 1.0% (w/v), the preferred range for each preservative, alone or in combination with others, is: benzyl alcohol (0.1-1.0%), or m-cresol (0.1-0.6%), or phenol (0.1-0.8%) or combination of methyl (0.05-0.25%) and ethyl or propyl or butyl (0.005%-0.03%) parabens.
  • the parabens are lower alkyl esters of para-hydroxybenzoic acid.
  • the crystalline trientine dihydrochloride salt may be administered parenterally (including subcutaneous injections, intravenous, intramuscular, intradermal injection or infusion techniques) or by inhalation spray in dosage unit formulations containing conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
  • sodium chloride or other salt may also be desirable to add sodium chloride or other salt to adjust the tonicity of the pharmaceutical formulation, depending on the tonicifier selected. However, this is optional and depends on the particular formulation selected. Parenteral formulations must be isotonic or substantially isotonic otherwise significant irritation and pain would occur at the site of administration.
  • the desired isotonicity may be accomplished using sodium chloride or other pharmaceutically acceptable agents such as dextrose, boric acid, sodium tartrate, propylene glycol, polyols (such as mannitol and sorbitol), or other inorganic or organic solutes.
  • the composition is isotonic with the blood of the subject.
  • the parenteral formulation may be thickened with a thickening agent such as methyl cellulose.
  • a thickening agent such as methyl cellulose.
  • the formulation may be prepared in an emulsified form, either water in oil or oil in water. Any of a wide variety of pharmaceutically acceptable emulsifying agents may be employed including, for example, acacia powder, a non-ionic surfactant or an ionic surfactant.
  • aqueous suspensions such as synthetic and natural gums i.e. tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin.
  • Water of suitable quality for parenteral administration must be prepared either by distillation or by reverse osmosis. Only by these means is it possible to separate adequately various liquid, gas and solid contaminating substances from water.
  • Water for injection is the preferred aqueous vehicle for use in the pharmaceutical formulation of the present invention. The water may be purged with nitrogen gas to remove any oxygen or free radicals of oxygen from the water.
  • Such additional ingredients may include wetting agents, oils (e.g., a vegetable oil such as sesame, peanut or olive), analgesic agents, emulsifiers, antioxidants, bulking agents, tonicity modifiers, metal ions, oleaginous vehicles, proteins (e.g., human serum albumin, gelatin or proteins) and a zwitterion (e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine).
  • oils e.g., a vegetable oil such as sesame, peanut or olive
  • analgesic agents emulsifiers, antioxidants, bulking agents, tonicity modifiers, metal ions, oleaginous vehicles
  • proteins e.g., human serum albumin, gelatin or proteins
  • a zwitterion e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine.
  • Containers are also an integral part of the formulation of an injection and may be considered a component, for there is no container that is totally insoluble or does not in some way affect the liquid it contains, particularly if the liquid is aqueous. Therefore, the selection of a container for a particular injection must be based on a consideration of the composition of the container, as well as of the solution, and the treatment to which it will be subjected.
  • each vial is sealed with a rubber closure held in place by an aluminum band.
  • Stoppers for glass vials such as, West 4416/50, 4416/50 (Teflon faced) and 4406/40, Abbott 5139 or any equivalent stopper can be used as the closure for the dose vial. These stoppers pass the stopper integrity test when tested using patient use patterns, e.g., the stopper can withstand at least about 100 injections.
  • the manufacturing process for the above formulation involves compounding, sterile filtration and filling steps.
  • the compounding procedure may for example, involve the dissolution of ingredients in a specific order, such as the preservative first followed by the stabilizer/tonicity agents, buffers and then the trientine active agent or dissolving all of the ingredients forming the parenteral formulation at the same time.
  • An example of one method of preparing a parenteral formulation for administration is the dissolution of the trientine active form, for example, trientine hydrochloride, in water and diluting the resultant mixture to 154 mM in a phosphate buffered saline.
  • parenteral formulations of the present invention are prepared by mixing the ingredients following generally accepted procedures.
  • the selected components may be mixed in a blender or other standard device to produce a concentrated mixture which may then be adjusted to the final concentration and viscosity by the addition of water, a thickening agent, a buffer, 5% human serum albumin or an additional solute to control tonicity.
  • the trientine active agent can be packaged as a dry solid and/or powder to be reconstituted with a solvent to yield a parenteral formulation in accordance with the present invention for use at the time of reconstitution.
  • the manufacturing process may include any suitable sterilization process when developing the parenteral formulation of the present invention.
  • Typical sterilization processes include filtration, steam (moist heat), dry heat, gases (e.g., ethylene oxide, formaldehyde, chlorine dioxide, propylene oxide, beta-propiolacctone, ozone, chloropicrin, peracetic acid methyl bromide and the like), radiant exposure and aseptic handling.
  • Suitable routes of parenteraladministration include intramuscular, intravenous, subcutaneous, intradermal, intraarticular, intrathecal and the like.
  • the subcutaneous route of administration is preferred. Mucosal delivery is also permissible.
  • the dose and dosage regimen will depend upon the weight and health of the subject.
  • Routes for parenteral administration therefore include intravenous, intramuscular, intraperitoneal, sub dermal, and subcutaneous administration.
  • the rate and duration of drug delivery may be controlled by, for example by using mechanically controlled drug infusion pumps.
  • the pharmaceutically acceptable active agent for example, one or more copper chelators, such as, for example, trientine or salts thereof such as trientine dihydrochloride, can be administered in the form of a depot injection that may be formulated in such a manner as to permit a sustained release of the active ingredient.
  • the active ingredient can be compressed into pellets or small cylinders and implanted subcutaneously or intramuscularly.
  • the pellets, or cylinders may additionally be coated with a suitable biodegradable polymer chosen so as to provide a desired release profile.
  • the active ingredient may alternatively be micropelleted.
  • Active agent micropellets using bioacceptable polymers can be designed to allow release rates to be manipulated to provide a desired release profile.
  • injectable depot forms can be made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations can also be prepared by entrapping the drug in liposomes, examples of which include unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearyl aamine or phosphatidylcholines. Depot injectable formulations can also be prepared by entrapping the drug in microemulsions which are compatible with body tissue.
  • phospholipids such as cholesterol, stearyl aamine or phosphatidylcholines.
  • Depot injectable formulations can also be prepared by entrapping the drug in microemulsions which are compatible with body tissue.
  • Implantable infusion devices may employ inert material such as biodegradable polymers listed above or synthetic silicones for example cylastic, silicone rubber or other polymers manufactured by the Dow-Corning Corporation.
  • the polymer may be loaded with active agent and any excipients.
  • Implantable infusion devices may also comprise a coating of, or a portion of, a medical device wherein the coating comprises the polymer loaded with active agent and any excipient.
  • Such an implantable infusion device may be prepared as disclosed in U.S. Pat. No.
  • An implantable infusion device may also be prepared by the in situ formation of an active agent containing solid matrix as disclosed in U.S. Pat. No. 6,120,789, herein incorporated in its entirety.
  • Implantable infusion devices may be passive or active.
  • An active implantable infusion device may comprise an active agent reservoir, a means of allowing the active agent to exit the reservoir, for example a permeable membrane, and a driving force to propel the active agent from the reservoir.
  • Such an active implantable infusion device may additionally be activated by an extrinsic signal, such as that disclosed in WO 02/45779, wherein the implantable infusion device comprises a system configured to deliver the active agent comprising an external activation unit operable by a user to request activation of the implantable infusion device, including a controller to reject such a request prior to the expiration of a lockout interval.
  • an active implantable infusion device include implantable drug pumps.
  • Implantable drug pumps include, for example, miniature, computerized, programmable, refillable drug delivery systems with an attached catheter that inserts into a target organ system, usually the spinal cord or a vessel. See Medtronic Inc.
  • Implantable drug infusion pumps are indicated for long-term intrathecal infusion of morphine sulfate for the treatment of chronic intractable pain; intravascular infusion of floxuridine for treatment of primary or metastatic cancer; intrathecal injection (baclofen injection) for severe spasticity; long-term epidural infusion of morphine sulfate for treatment of chronic intractable pain; long-term intravascular infusion of doxorubicin, cisplatin, or methotrexate for the treatment or metastatic cancer; and long-term intravenous infusion of clindamycin for the treatment of osteomyelitis.
  • Such pumps may also be used for the long-term infusion of one or more copper chelators, for example, for example, trientine or salts thereof, at a desired amount for a desired number of doses or steady state administration.
  • One form of a typical implantable drug infusion pump (Synchromed EL programmable pump; Medtronic) is titanium covered and roughly disk shaped, measures 85.2 mm in diameter and 22.86 mm in thickness, weighs 185 g, has a drug reservoir of 10 mL, and runs on a lithium thionyl-chloride battery with a 6- to 7-year life, depending on use.
  • the downloadable memory contains programmed drug delivery parameters and calculated amount of drug remaining, which can be compared with actual amount of drug remaining to access accuracy of pump function, but actual pump function over time is not recorded.
  • the pump is usually implanted in the right or left abdominal wall.
  • Other pumps useful in the invention include, for example, portable disposable infuser pumps (PDIPs).
  • PDIPs portable disposable infuser pumps
  • implantable infusion devices may employ liposome delivery systems such as a small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles can be formed from a variety of phospholipids, such as cholesterol, stearyl amine or phosphatidylcholines.
  • the invention also includes delayed-release ocular preparations containing one or more copper chelators, for example, trientine or salts thereof.
  • Trientine therapy is effective in treating diabetic arterial disease.
  • This aspect of the invention provides ocular preparations of trientine suitable for administration to humans for the treatment of the disease of the retinal arteries in diabetes. Such administration is expected to yield high, localized concentrations of drug, suitable for treatment of diabetic arterial disease in the retina, and diabetic retinopathy.
  • ophthalmic solutions One of the problems associated with the use of ophthalmic solutions is the rapid loss of administered drug due to blinking of the eye and the flushing effect of lacrimal fluids. Up to 80% of an administered dose may be lost through tears and the action of nasolacrimal drainage within 5 minutes of installation. Extended periods of therapy may be achieved by formulations that increase the contact time between the medication and the corneal surface. This may be accomplished through use of agents that increase the viscosity of solutions; by ophthalmic suspensions in which the drug particles slowly dissolve; by slowly dissipating ophthalmic ointments; or by use of ophthalmic inserts.
  • Preparations of one or more copper chelators for example, trientine or its salts suitable for ocular administration to humans may be formulated using synthetic high molecular weight cross-linked polymers such as those of acrylic acid (e.g., Carbopol 940) or gellan gum (Gelrite; see, Merck Index 12th Ed., 4389), a compound that forms a gel upon contact with the precorneal tear film (e.g. as employed in Timoptic-XE by Merck, Inc.).
  • synthetic high molecular weight cross-linked polymers such as those of acrylic acid (e.g., Carbopol 940) or gellan gum (Gelrite; see, Merck Index 12th Ed., 4389), a compound that forms a gel upon contact with the precorneal tear film (e.g. as employed in Timoptic-XE by Merck, Inc.).
  • Further embodiments include delayed-release ocular preparations containing trientine in ophthalmic inserts, such as the OCUSERT system (Alza Inc.).
  • ophthalmic inserts such as the OCUSERT system (Alza Inc.).
  • OCUSERT system Alza Inc.
  • inserts are elliptical with dimensions of about 13.4 mm by 5.4 mm by 0.3 mm (thickness).
  • the insert is flexible and has a drug-containing core surrounded on each side by a layer of hydrophobic ethylene/vinyl acetate copolymer membranes through which the drug diffuses at a constant rate.
  • the white margin around such devices contains white titanium dioxide, an inert compound that confers visibility.
  • the rate of drug diffusion is controlled by the polymer composition, the membrane thickness, and the drug solubility.
  • the drug-containing inserts may be placed in the conjunctival sac from which they release their medication over a typical 7-d period in the treatment of diabetic retinal disease.
  • Another form of an ophthalmic insert is a rod shaped, water soluble structure composed of hydroxypropyl cellulose in which trientine is embedded. The insert is placed into the inferior cul-de-sac of the eye once or twice daily in the treatment of diabetic retinal disease. The inserts soften and slowly dissolve, releasing the drug that is then taken up by the ocular fluids.
  • Lacrisert Merck Inc.
  • the invention also provides in part dose delivery formulations and devices formulated to enhance bioavailability of trientine active agent. This may be in addition to or in combination with any of the dose delivery formulations or devices described above.
  • trientine is poorly absorbed in the digestive tract and consequently its bioavailability is incomplete, and may be irregular or vary from one person to another.
  • a therapeutically effective amount of trientine active agent is an amount capable of providing an appropriate level of trientine active agent in the bloodstream. By increasing the bioavailability of trientine active agent, a therapeutically effective level of trientine active agent may be achieved by administering lower dosages than would otherwise be necessary.
  • An increase in bioavailability of trientine active agent may be achieved by complexation of trientine active agent with one or more bioavailability or absorption enhancing agents or in bioavailability or absorption enhancing formulations.
  • the invention in part provides for the formulation of trientine active agent with other agents useful to enhance bioavailability or absorption.
  • bioavailability or absorption enhancing agents include, but are not limited to, various surfactants such as various triglycerides, such as from butter oil, monoglycerides, such as of stearic acid and vegetable oils, esters thereof, esters of fatty acids, propylene glycol esters, the polysorbates, sodium lauryl sulfate, sorbitan esters, sodium sulfosuccinate, among other compounds.
  • cyclodextrin examples include carrier molecules such as cyclodextrin and derivatives thereof, well known in the art for their potential as complexation agents capable of altering the physicochemical attributes of drug molecules.
  • cyclodextrin may stabilize (both thermally and oxidatively), reduce the volatility of, and alter the solubility of, active agents with which they are complexed.
  • Cyclodextrins are cyclic molecules composed of glucopyranose ring units which form toroidal structures. The interior of the cyclodextrin molecule is hydrophobic and the exterior is hydrophilic, making the cyclodextrin molecule water soluble.
  • the degree of solubility can be altered through substitution of the hydroxyl groups on the exterior of the cyclodextrin.
  • the hydrophobicity of the interior can be altered through substitution, though generally the hydrophobic nature of the interior allows accommodation of relatively hydrophobic guests within the cavity.
  • Accommodation of one molecule within another is known as complexation and the resulting product is referred to as an inclusion complex.
  • examples of cyclodextrin derivatives include sulfobutylcyclodextrin, maltosylcyclodextrin, hydroxypropylcyclodextrin, and salts thereof.
  • Complexation of trientine with a carrier molecule such as cyclodextrin to form an inclusion complex may thereby reduce the size of the trientine dose needed for therapeutic efficacy by enhancing the bioavailability of the administered trientine.
  • a microemulsion is a fluid and stable homogeneous solution composed of four major constituents, respectively, a hydrophilic phase, a lipophilic phase, at least one surfactant (SA) and at least one cosurfactant (CoSA).
  • SA surfactant
  • CoSA cosurfactant
  • a surfactant is a chemical compound possessing two groups, the first polar or ionic, which has a great affinity for water, the second which contains a longer or shorter aliphatic chain and is hydrophobic. These chemical compounds having marked hydrophilic character are intended to cause the formation of micelles in aqueous or oily solution.
  • Suitable surfactants include mono-, di- and triglycerides and polyethylene glycol (PEG) mono- and diesters.
  • a cosurfactant also sometimes known as “co-surface-active agent”, is a chemical compound having hydrophobic character, intended to cause the mutual solubilization of the aqueous and oily phases in a microemulsion.
  • suitable co-surfactants include ethyl diglycol, lauric esters of propylene glycol, oleic esters of polyglycerol, and related compounds.
  • the invention in part provides for the formulation of trientine active agent with various polymers to enhance bioavailability by increasing adhesion to mucosal surfaces, by decreasing the rate of degradation by hydrolysis or enzymatic degradation of the active agent, and by increasing the surface area of the active agent relative to the size of the particle.
  • Suitable polymers can be natural or synthetic, and can be biodegradable or non-biodegradable. Delivery of low molecular weight active agents such as trientine active agent may occur by either diffusion or degredation of the polymeric system.
  • Representative natural polymers include proteins such as zein, modified zein, casein, gelatin, gluten, serum albumin, and collagen, polysaccharides such as cellulose, dextrans, and polyhyaluronic acid.
  • Synthetic polymers are generally preferred due to the better characterization of degradation and release profiles.
  • Representative synthetic polymers include polyphosphazenes, poly(vinyl alcohols), polyamides, polycarbonates, polyacrylates, polyalkylenes, polyacrylamides, polyalkylene glycols, polyalkylene oxides, polyalkylene terephthalates, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof.
  • polyacrylates examples include poly(methyl methacrylate), poly(ethyl methacrylate), poly(butyl methacrylate), poly(isobutyl methacrylate), poly(hexyl methacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate) and poly(octadecyl acrylate).
  • Synthetically modified natural polymers include cellulose derivatives such as alkyl celluloses, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, and nitrocelluloses.
  • Suitable cellulose derivatives include methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxymethyl cellulose, cellulose triacetate and cellulose sulfate sodium salt.
  • Each of the polymers described above can be obtained from commercial sources such as Sigma Chemical Co., St. Louis, Mo., Polysciences, Warrenton, Pa., Aldrich Chemical Co., Milwaukee, Wis., Fluka, Ronkonkoma, N.Y., and BioRad, Richmond, Calif. or can be synthesized from monomers obtained from these suppliers using standard techniques.
  • polymers described above can be separately characterized as biodegradable, non-biodegradable, and bioadhesive polymers, as discussed in more detail below.
  • Representative synthetic degradable polymers include polyhydroxy acids such as polylactides, polyglycolides and copolymers thereof, poly(ethylene terephthalate), poly(butic acid), poly(valeric acid), poly(lactide-co-caprolactone), polyanhydrides, polyorthoesters and blends and copolymers thereof
  • Representative natural biodegradable polymers include polysaccharides such as alginate, dextran, cellulose, collagen, and chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), and proteins such as albumin, zein and copolymers and blends thereof, alone or in combination with synthetic polymers.
  • non-biodegradable polymers include ethylene vinyl acetate, poly(meth)acrylic acid, polyamides, polyethylene, polypropylene, polystyrene, polyvinyl chloride, polyvinylphenol, and copolymers and mixtures thereof.
  • Hydrophilic polymers and hydrogels tend to have bioadhesive properties.
  • Hydrophilic polymers that contain carboxylic groups e.g., poly[acrylic acid]
  • Polymers with the highest concentrations of carboxylic groups are preferred when bioadhesiveness on soft tissues is desired.
  • cellulose derivatives such as sodium alginate, carboxymethylcellulose, hydroxymethylcellulose and methylcellulose also have bioadhesive properties. Some of these bioadhesive materials are water-soluble, while others are hydrogels.
  • Polymers such as hydroxypropylmethylcellulose acetate succinate (HPMCAS), cellulose acetate trimellitate (CAT), cellulose acetate phthalate (CAP), hydroxypropylcellulose acetate phthalate (HPCAP), hydroxypropylmethylcellulose acetate phthalate (HPMCAP), and methylcellulose acetate phthalate (MCAP) may be utilized to enhance the bioavailibity of drugs with which they are complexed.
  • HPMCAS hydroxypropylmethylcellulose acetate succinate
  • CAT cellulose acetate trimellitate
  • CAP cellulose acetate phthalate
  • HPCAP hydroxypropylcellulose acetate phthalate
  • MCAP methylcellulose acetate phthalate
  • Rapidly bioerodible polymers such as poly(lactide-co-glycolide), polyanhydrides, and polyorthoesters, whose carboxylic groups are exposed on the external surface as their smooth surface erodes, can also be used for bioadhesive drug delivery systems.
  • polymers containing labile bonds such as polyanhydrides and polyesters, are well known for their hydrolytic reactivity. Their hydrolytic degradation rates can generally be altered by simple changes in the polymer backbone. Upon degradation, these materials also expose carboxylic groups on their external surface, and accordingly, these can also be used for bioadhesive drug delivery systems.
  • agents that may enhance bioavailability or absorption can act by facilitating or inhibiting transport across the intestinal mucosa
  • agents that increase blood flow such as vasodilators, may increase the rate of absorption of orally administered drugs by increasing the blood flow to the gastrointestinal tract.
  • Vasodilators have been used in combination with other drugs.
  • a coronary vasodilator diltiazem
  • drugs which have an absolute bioavailability of not more than 20%, such as adrenergic beta-blocking agents (e.g., propranolol), catecholamines (e.g., dopamine), benzodiazepine derivatives (e.g., diazepam), vasodilators (e.g., isosorbide dinitrate, nitroglycerin or amyl nitrite), cardiotonics or antidiabetic agents, bronchodilators (e.g., tetrahydroisoquinoline), hemostatics (e.g., carbazochrome sulfonic acid), antispasmodics (e.g., timepidium halide) and antitussives (e.g., tipepidine).
  • Vasodilators therefore constitute another class
  • Other mechanisms of enhancing bioavailability of the compounds and formulations of the invention include the inhibition of reverse active transport mechanisms.
  • one of the active transport mechanisms present in the intestinal epithelial cells is p-glycoprotein transport mechanism which facilitates the reverse transport of substances, which have diffused or have been transported inside the epithelial cell, back into the lumen of the intestine.
  • the p-glycoprotein present in the intestinal epithelial cells may function as a protective reverse pump which prevents toxic substances which have been ingested and diffused or transported into the epithelial cell from being absorbed into the circulatory system and becoming bioavailable.
  • the p-glycoprotein in the intestinal cell can also function to prevent bioavailability of substances which are beneficial, such as certain drugs which happen to be substrates for the p-glycoprotein reverse transport system. Inhibition of this p-glycoprotein mediated active transport system will cause less drug to be transported back into the lumen and will thus increase the net drug transport across the gut epithelium and will increase the amount of drug ultimately available in the blood.
  • p-glycoprotein inhibitors are well known and appreciated in the art.
  • inhibition of a reverse active transport system of which, for example, a trientine active agent is a substrate may thereby enhance the bioavailability of said trientine active agent.
  • trientine dihydrochloride is effective at removing Cu from diabetic rats at doses far lower than have been previously shown to be effective.
  • FIG. 3 and particularly in FIG. 4 which presents Cu excretion normalised to body weight, Cu excretion in the urine of diabetic rats administered trientine at a dose of 0.1 mg.kg ⁇ 1 (the lowest dose administered in the studies presented herein) is significantly increased over that of diabetic rats administered saline.
  • trientine active agents including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on, will be effective at doses lower than, for example, the 1.2 g.d ⁇ 1 herein shown to be effective in treating human heart disease. It may be effective at doses in the order of 1/10, 1/100 and even 1/1000 of those we have already employed (e.g. in the order of 120 mg.d ⁇ 1 , 12 mg.d ⁇ 1 or even 1.2 mg.d ⁇ 1 ).
  • the invention accordingly in part provides low-dose dose delivery formulations and devices comprising one or more trientine active agents, including but not limited to trientine, trientine salts, trientine analogues of formulae I and II, and so on, in an amount sufficient to provide, for example, dosage rates from 0.01 mg.kg ⁇ 1 to 5 mg.kg ⁇ 1 , 0.01 mg.kg ⁇ 1 to 4.5 mg.kg ⁇ 1 , 0.02 mg.kg ⁇ 1 to 4 mg.kg ⁇ 1 , 0.02 to 3.5 mg.kg ⁇ 1 , 0.02 mg.kg ⁇ 1 to 3 mg.kg ⁇ 1 , 0.05 mg.kg ⁇ 1 to 2.5 mg.kg ⁇ 1 , 0.05 mg.kg ⁇ 1 to 2 mg.kg ⁇ 1 , 0.05-0.1 mg.kg ⁇ 1 to 5 mg.kg ⁇ 1 , 0.05-0.1 mg.kg ⁇ 1 to 4 mg.kg ⁇ 1 , 0.05-0.1 mg.kg ⁇ 1 to 3 mg.kg ⁇ 1 ,
  • any such dose may be administered by any of the routes or in any of the forms herein described. It will be appreciated that any of the dose delivery formulations or devices described herein particularly for oral administration may be utilized, where applicable or desirable, in a dose delivery formulation or device for administration by any of the other routes herein contemplated or commonly employed. For example, it could be given parenterally using a dose form suitable for parenteral administration, or be delivered in an oral dosage form such as a modified release, extended release, delayed release, slow release or repeat action oral dosage form.
  • Another aspect of the invention base on results of studies described herein that equate human copper values depletion against those of the STZ rat, a dosage form each with less than 250 mg of trientine dihydrochloride (or trientine active agent when expressed as the dihydrochloride). Envisaged are capsule forms having less than 250 mg trientine dihydrochloride or equivalent thereof of trientine active agent per capsule or tablets or capsules of any suitable form.
  • At risk refers to mammals subjected to a risk assessment of a kind exemplified in the Journal of American Medical Association, May 16, 2001, Volume 285 No. 19, 2486-2497 where Framingham risk scoring which takes account of age, total cholesterol, HDL cholesterol, systolic blood pressure, treatment for hypertension and cigarette smoking is mentioned and to which can be added glucose abnormalities of any of the kinds herein described.
  • This Example was carried out to determine for the sake of subsequent comparison baseline physiological data relating to the effects of streptozotocin (STZ) treatment in rats, in addition to baseline physiological data from diabetic and nondiabetic rats.
  • STZ streptozotocin
  • streptozotocin STZ, 55 mg.kg ⁇ 1 body weight, Sigma; St. Louis, Mo.
  • both diabetic and nondiabetic rats were housed in like-pairs and provided with access to normal rat chow (Diet 86 pellets; New Zealand Stock Feeds, Auckland, NZ) and deionized water ad libitum. Blood glucose and body weight were measure at day 3 following STZ/saline injection and then weekly throughout the study. Diabetes was identified by polydipsia, polyuria and hyperglycemia (>11 mmol.1 ⁇ 1 , Advantage II, Roche Diagnostics, NZ Ltd).
  • Results were as follows. With regard to Effects of STZ on blood glucose and body weight, blood glucose increased to 25 ⁇ 2 mmol.1 ⁇ 1 three days following STZ injection (Table 1). Despite a greater daily food intake, diabetic animals lost weight whilst nondiabetic animals continued to gain weight during the 44 days following STZ/saline injection. On the day of the experiment blood glucose levels were 24 ⁇ 1 and 5 ⁇ 0 mmol.1 ⁇ 1 and body weight 264 ⁇ 7 g and 434 ⁇ 9 g for diabetic and nondiabetic animals respectively.
  • This Example assessed the effect of acute intravenous administration of increasing doses of trientine on the excretion profiles of copper and iron in the urine of diabetic and nondiabetic rats.
  • the trachea was cannulated and the animal ventilated at 70-80 breaths.min ⁇ 1 with air supplemented with O2 (Pressure Controlled Ventilator, Kent Scientific, Connecticut, USA).
  • O2 Pressure Controlled Ventilator, Kent Scientific, Connecticut, USA.
  • the respiratory rate and end-tidal pressure (10-15 cmH2O) were adjusted to maintain end-tidal CO2 at 35-40 mmHg (SC-300 CO2 monitor, Pryon Corporation, Wisconsin, USA).
  • Body temperature was maintained at 37° C. throughout surgery and the experiment by a heating pad.
  • Estimated fluid loss was replaced with intravenous administration of 154 mmol.1 ⁇ 1 NaCl solution at a rate of 5 ml.kg ⁇ 1 .h ⁇ 1 .
  • Trientine was administered intravenously over 60 s in hourly doses of increasing concentration (0.1, 1.0, 10 and 100 mg.kg ⁇ 1 in 75 ⁇ l saline followed by 125 ⁇ l saline flush). Control animals received an equivalent volume of saline.
  • Urine was collected in 15 min aliquots throughout the experiment in pre-weighed polyethylene epindorf tubes. At the end of the experiment a terminal blood sample was taken by cardiac puncture and the separated serum stored at ⁇ 80° C. until future analysis. Hearts were removed through a rapid mid-sternal thoracotomy and processed as described below.
  • MAP Mean arterial pressure
  • HR heart rate
  • HR heart rate
  • HR heart rate
  • HR heart rate
  • HR heart rate
  • core body temperature core body temperature
  • Urine and tissue analysis was carried out as follows. Instrumentation: A Perkin Elmer (PE) Model 3100 Atomic Absorption Spectrophotometer equipped with a PE HGA-600 Graphite Furnace and PE AS-60 Furnace Autosampler was used for Cu and Fe determinations in urine. Deuterium background correction was employed. A Cu or Fe hollow-cathode lamp (Perkin Elmer Corporation) was used and operated at either 10 W (Cu) or 15 W (Fe). The 324.8 nm atomic line was used for Cu and the 248.3 nm atomic line for Fe. The slit width for both Cu and Fe was 0.7 nm. Pyrolytically coated graphite tubes were used for all analyses. The injection volume was 20 ⁇ L.
  • a typical graphite furnace temperature program is shown below: GF-AAS temperature program Procedure Temp/° C. Ramp/s Hold/s Int. Flow/mL min ⁇ 1 Drying 90 1 5 300 120 60 5 300 Pre-treatment 1250* 20 10 300 20 1 10 300 Atomization - 2300/2500 1 8 0 Cu/Fe Post-treatment 2600 1 5 300 *A pre-treatment temperature of 1050° C. was used for tissue digest analyses (see Example 3)
  • Reagents All reagents used were of the highest purity available and at least of analytical grade.
  • GF-AAS standard working solutions of Cu and Fe were prepared by stepwise dilution of 1000 mg.1 ⁇ 1 (Spectrosol standard solutions; BDH). Water was purified by a Millipore Milli-Q ultra-pure water system to a resistivity of 18 M ⁇ .
  • Urine Urine was collected in pre-weighed 1.5 ml micro test tubes (eppendorf). After reweighing, the urine specimens were centrifuged and the supernatant diluted 25:1 with 0.02 M 69% Aristar grade HNO 3 . The sample was stored at 4° C. prior to GF-AAS analysis. If it was necessary to store a sample for a period in excess of 2 weeks, it was frozen and kept at ⁇ 20° C.
  • Serum Terminal blood samples were centrifuged and serum treated and stored as per urine until analysis.
  • diabetic animals consistently excreted significantly more urine than nondiabetic animals except in response to the highest dose of drug (100 mg.kg ⁇ 1 ) or equivalent volume of saline ( FIG. 16 ).
  • Administration of the 100 mg.kg ⁇ 1 dose of trientine also increased urine excretion in nondiabetic animals to greater than that of nondiabetic animals receiving the equivalent volume of saline ( FIG. 17 ). This effect was not seen in diabetic animals.
  • Electron paramagnetic resonance spectroscopy showed that the urinary Cu from drug-treated animals was mainly complexed as trientine-Cu II ( FIG. 28 ), indicating that the increased tissue Cu in diabetic rats is mainly divalent. These data indicate that rats with severe hyperglycaemia develop increased systemic Cu II that can be extracted by selective chelation.
  • This example was carried out to determine the effect of acute intravenous administration of increasing doses of trientine on the copper and iron content of cardiac tissue in normal and diabetic rates.
  • Standard Reference Material 1577b Bovine Liver was obtained from the National Institute of Standards and Technology and used to evaluate the efficiency of tissue digestion. The results obtained are reported below: GF-AAS and ICP-MS results for NIST SRM 1577b bovine liver* Element Certified value GF-AAS ICP-MS Cu 160 ⁇ 8 142 ⁇ 12 164 ⁇ 12 Fe 184 ⁇ 15 182 ⁇ 21 166 ⁇ 14 Zn 127 ⁇ 16 — 155 ⁇ 42 *Measured in ⁇ g ⁇ g ⁇ 1 of dry matter.
  • Sample pre-treatment was carried out as follows. Heart: Following removal from the animal, the heart was cleaned of excess tissue, rinsed in buffer to remove excess blood, blotted dry and a wet ventricular weight recorded. Using titanium instruments a segment of left ventricular muscle was dissected and placed in a pre-weighed 5.0 ml polystyrene tube. The sample was freeze-dried overnight to constant weight before 0.45 ml of 69% Aristar grade HNO 3 was added. The sample tube was heated in a water bath at 65° C. for 60 minutes. The sample was brought to 4.5 ml with Milli-Q H 2 O. The resulting solution was diluted 2:1 in order to reduce the HNO 3 concentration below the maximum permitted for ICP-MS analysis.
  • diabetic rats excreted significantly higher levels of copper across all dose levels (see FIG. 27 ).
  • Baseline copper excretion was also significantly higher in diabetic rats compared to nondiabetic rats. There was no difference at baseline levels between the drug and saline groups.
  • the interaction effect for the model was significant at dose levels of 1.0 mg.kg ⁇ 1 and above. The presence of a significant interaction term means that the influence of one effect varies with the level of the other effect. Therefore, the outcome of a significant interaction between the diabetes and drug factors is increased copper excretion above the predicted additive effects of these two factors.
  • Trientine treatment effectively increases copper excretion in both diabetic and nondiabetic animals.
  • the excretion of copper in urine following trientine administration is greater per gram of bodyweight in diabetic than in nondiabetic animals. Iron excretion was not increased by trientine treatment in either diabetic or nondiabetic animals.
  • This experiment was carried out to compare cardiac function in trientine-treated and non-treated, STZ diabetic and normal rats using an isolated-working-rodent heart model.
  • STZ intravenous streptozotocin
  • All rats were given a short inhalational anesthetic (Induction: 5% halothane and 2 L/min oxygen, maintained on 2% halothane and 2 L/min oxygen).
  • STZ intravenous streptozotocin
  • Those in the two diabetic groups then received a single intravenous bolus dose of STZ (57 mg/kg body weight) in 0.5 ml of 0.9% saline administered via a tail vein.
  • Non-diabetic sham-treated animals received an equivalent volume of 0.9% saline.
  • Diabetic and non-diabetic rats were housed in like-pairs and provided with free access to normal rat chow (Diet 86 pellets; New Zealand Stock Feeds, Auckland, NZ) and deionized water ad libitum. Each cage had two water bottles on it to ensure equal access to water or drug for each animal. Animals were housed at 21 degrees and 60% humidity in standard rat cages with a sawdust floor that was changed daily.
  • Blood glucose was measured in tail-tip capillary blood samples (Advantage II, Roche Diagnostics, NZ Ltd). Sampling was performed on all groups at the same time of the day. Blood glucose and body weight were measured on day 3 following STZ/saline injection and then weekly throughout the study. Diabetes was confirmed by presence of polydipsia, polyuria and hyperglycemia (>11 mmol.L ⁇ 1 ).
  • trientine was prepared in the drinking water for each cage at a concentration of 50 mg/L.
  • the trientine-containing drinking water was administered continuously from the start of week 7 until the animal was sacrificed at the end of week 13.
  • the drug concentration in their drinking water was adjusted so that they consumed approximately the same dose as the corresponding STZ/D7 group.
  • Trientine treated animals ingested mean drug doses of between 8 to 11 mg per day.
  • mice were anesthetized (5% halothane and 2 L.min ⁇ 1 O 2 ), and heparin (500 IU.kg ⁇ 1 ) (Weddel Pharmaceutical Ltd., London) administered intravenously via tail vein.
  • heparin 500 IU.kg ⁇ 1
  • a 2 ml blood sample was then taken from the inferior vena cava and the heart was then rapidly excised and immersed in ice-cold Krebs-Henseleit bicarbonate buffer to arrest contractile activity. Hearts were then placed in the isolated perfused working heart apparatus.
  • the aortic root of the heart was immediately ligated to the aortic cannula of the perfusion apparatus.
  • Retrograde (Langendorff) perfusion at a hydrostatic pressure of 100 cm H 2 O and at 37° C. was established and continued for 5 min while cannulation of the left atrium via the pulmonary vein was completed.
  • the non-working (Langendorff) preparation was then converted to the working heart model by switching the supply of perfusate buffer from the aorta to the left atrium at a filling pressure of 10 cm H 2 O.
  • the left ventricle spontaneously ejected into the aortic cannula against a hydrostatic pressure (after-load) of 76 cmH 2 O (55.9 mmHg).
  • the perfusion solution was Krebs-Henseleit bicarbonate buffer (mM: KCl 4.7, CaCl 2 2.3, KH 2 PO 4 1.2, MgSO 4 1.2, NaCl 118, and NaHCO 3 25), pH 7.4 containing 11 mM glucose and it was continuously gassed with 95% O 2 :5% CO 2 .
  • the buffer was also continuously filtered in-line (initial 8 ⁇ m, following 0.4 ⁇ m cellulose acetate filters; Sartorius, Germany).
  • the temperature of the entire perfusion apparatus was maintained by water jackets and buffer temperature was continuously monitored and adjusted to maintain hearts at 37° C. throughout perfusion.
  • a modified 24 g plastic intravenous cannula (Becton Dickson, Utah, USA) was inserted into the left ventricle via the apex of the heart using the normal introducer-needle. This cannula was subsequently attached to a SP844 piezo-electric pressure transducer (AD Instruments) to continuously monitor left ventricular pressure. Aortic pressure was continuously monitored through a side arm of the aortic cannula with a pressure transducer (Statham Model P23XL, Gould Inc., Calif., USA).
  • the heart was paced (Digitimer Ltd, Heredfordshire, England) at a rate of 300 bpm by means of electrodes attached to the aortic and pulmonary vein cannulae using supra-threshold voltages with pulses of 5-ms duration from the square wave generator.
  • Aortic flow was recorded by an in-line flow meter (Transonic T206, Ithaca, N.Y., USA) and coronary flow was measured by timed 30 sec collection of the coronary vein effluent at each time point step of the protocol.
  • the working heart apparatus used was a variant of that originally described by Neely, J R, et al., Am J Physiol 212:804-14 (1967).
  • the modified apparatus allowed measurements of cardiac function at different pre-load pressures ( FIG. 14 and FIG. 15 ). This was achieved by constructing the apparatus so that the inflow height of the buffer coming to the heart could be altered through a series of graduated steps in a reproducible manner. As in the case of the pre-load, the outflow tubing from the aorta could also be increased in height to provide a series of defined after-load pressures.
  • the after-load heights have been converted to mm Hg for presentation in the results which is in keeping with published convention.
  • the data processing functions of this device were used to calculate the first derivative of the two pressure waves (ventricular and aortic).
  • the final cardiac function data available comprised:
  • Cardiac output (CO) is the amount of buffer pumped per unit time by the heart and is comprised of buffer that is pumped out the aorta as well as the buffer pumped into the coronary vessels. This is an overall indicator of cardiac function.
  • +dP/dt is the rate of change of ventricular (or aortic pressure) and correlates well with the strength of the contraction of the ventricle (contractility). It can be used to compare contractility abilities of different hearts when at the same pre-load (Textbook of Medical Physiology, Ed. A. Guyton. Saunders company 1986). ⁇ dP/dt is an accepted measurement of the rate of relaxation of the ventricle].
  • the experiment was divided into two parts, the first with fixed after-load and variable pre-load the second, which immediately followed on from the first, with fixed pre-load and variable after-load.
  • the heart was initially allowed to equilibrate for 6 min at 10 cm H 2 O atrial filling pressure and 76 cm H 2 O after-load. During this period the left ventricular pressure transducer cannula was inserted and the pacing unit started. Once the heart was stable, the atrial filling pressure was then reduced to 5 cm H 2 O of water and then progressively increased in steps of 2.5 cmH 2 O over a series of 7 steps to a maximum of 20 cmH 2 O. The pre-load was kept at each filling pressure for 2 min, during which time the pressure trace could be observed to stabilize and the coronary flow was measured. On completion of the variable pre-load experiment, the variable after-load portion of the experiment was immediately commenced.
  • the filling pressure pre-load was set at 10 cm H 2 O and the after-load was then increased from 76 cm H 2 O (55.9 mm Hg) in 9 steps; of 2 min duration.
  • Data from the Powerlab was extracted by averaging 1 min intervals from the stable part of the electronic trace generated from each step in the protocol. The results from each group were then combined and analyzed for differences between the groups for the various cardiac function parameters (aortic flow, cardiac flow, MLVDP, LV or aortic ⁇ dP/dt). Differences between repeated observations at different pre-load conditions were explored and contrasted between study group using a mixed models approach to repeated measures (SAS v8.1, SAS Institute Inc, Cary N.C.). Missing random data were imputed using a maximum likelihood approach. Significant mean and interaction effects were further examined using the method of Tukey to maintain a pairwise 5% error rate for post hoc tests. All tests were two-tailed.
  • Results showing the weights of the animals at the end of the experimental period are found in Table 5. Diabetic animals were about 50% smaller than their corresponding age matched normals. A graph of the percentage change in weight for each experimental group is found in FIG. 5 , wherein the arrow indicates the start of trientine treatment.
  • the following graphs of FIGS. 7 to 12 represent cardiac performance parameters of the animals (STZ diabetic; STZ diabetic+drug; and sham-treated controls) while undergoing increasing atrial filling pressure (5-20 cmH 2 O, pre-load) with a constant after-load of 75 cm H 2 O. All results are mean ⁇ sem. In each graph for clarity unless otherwise stated, only significant differences related to the STZ/D7 the other groups are shown:* indicates p ⁇ 0.05 for STZ v STZ/D7, # p ⁇ 0.05 for STZ/D7 v Sham/D7. Unless stated, STZ/D7 v Sham or Sham/D7 was not significant.
  • Cardiac output ( FIG. 7 ) is the sum to the aortic flow ( FIG. 10 ) and the coronary flow as displayed in FIG. 8 . Since the control hearts and experimental groups have significantly different final weights, the coronary flow is also presented ( FIG. 9 ) as the flow normalized to heart weight (note that coronary flow is generally proportional to cardiac muscle mass, and therefore to cardiac weight).
  • the first derivative of the pressure curve gives the rate of change in pressure development in the ventricle with each cardiac cycle and the maximum positive rate of change (+dP/dt) value is plotted in FIG. 11 .
  • the corresponding maximum rate of relaxation ( ⁇ dP/dt) is in FIG. 12 . Similar results showing improvement in cardiac function were found from the data derived from the aortic pressure cannula (results not shown).
  • This Example was carried out to further evaluate the effect of acute trientine administration on cardiac tissue by assessing left ventricular (LV) histology.
  • LV histology was studied by laser confocal (LCM; FIG. 29 a - d ) and transmission electron microscopy (TEM; FIG. 29 e - h ).
  • LCM laser confocal
  • TEM transmission electron microscopy
  • LV sections were co-stained with phalloidin to visualize actin filaments, and ⁇ 1 -integrin as a marker for the extracellular space.
  • Ding B, et al. “Left ventricular hypertrophy in ascending aortic stenosis in mice: anoikis and the progression to early failure,” Circulation 101:2854-2862 (2000).
  • LCM LCM-LV sections were fixed (4% paraformaldehyde, 24 h); embedded (6% agar); vibratomed (120 pm, Campden); stained for f-actin (Phafloidin-488, molecular Probes) and ⁇ 1 -integrin antibody with a secondary antibody of goat anti-rabbit conjugated to CY5 (1:200; Ding B, et al., “Left ventricular hypertrophy in ascending aortic stenosis in mice: anoikis and the progression to early failure,” Circulation 101:2854-2862 (2000)); and visualised (TCS-SP2, Leica).
  • specimens were post-fixed (1:1 v/v 1% w/v 0s0 M 0s0 M PBS); stained (aqueous uranyl acetate (2% w/v, 20 mm) then lead citrate (3 mm)); sectioned (70 nm); and visualized (CM-12, Phillips).
  • TEM was largely consistent with LCM.
  • diabetes caused unmistakable myocardial damage characterized by loss of myocytes with evident myocytolysis; disorganization of remaining cardiomyocytes in which swollen mitochondria were prominent; and marked expansion of the extracellular space ( FIG. 29 f ).
  • Jackson C V, et al. “A functional and ultrastructural analysis of experimental diabetic rat myocardium: manifestation of acardiomyopathy,” Diabetes 34:876-883 (1985).
  • Oral trientine caused substantive recovery of LV structure in diabetics, with increased numbers and normalized orientation of myocytes; return to normal of mitochondrial structure; and marked narrowing of the extracellular space ( FIG.
  • the improvement restored function to such an extent that there was no significant difference between the drug treated and the sham-treated control groups; (5) The aortic transducer measures of pressure change also showed improved function in the drug treated diabetic group compared to the untreated diabetics (data not shown); (6) When after-load was increased in the presence of constant pre-load, it was observed that the heart's ability to function at higher after-loads was greatly improved in the drug treated diabetic group compared to the untreated diabetic group.
  • This Example was carried out to assess the effect of chronic trientine administration on cardiac structure and function in diabetic and non-diabetic humans.
  • Subjects (30-70 y) who provided written informed consent were eligible for inclusion if they had:T2DM with HbA 1c >7%; cardiac ejection fraction (echocardiography) ⁇ 45% with evidence of diastolic dysfunction but no regional wall-motion anomalies; no new medications for more than 6 months with no change of ⁇ -blocker dose; normal electrocardiogram (sinus rhythm, normal PR Interval, normal T wave and QRS configuration, and isoelectric ST segment); and greater than 90% compliance with single-blinded placebo therapy during a 2-w run-in period. Women were required to be post-menopausal, surgically sterile, or non-lactating and non-pregnant and using adequate contraception.
  • LV diastolic filling was assessed using mitral inflow Doppler (with pre-load reduction) to ensure patients had abnormalities of diastolic filling; no patient with normal mitral filling proceeded to randomisation.
  • Subjects meeting inclusion criteria and with no grounds for exclusion were then randomised to receive trientine (600 mg twice-daily) before meals (total dose 1.2 g.d ⁇ 1 ) or 2 identical placebo capsules twice-daily before meals, in a double-blind, parallel-group design.
  • Treatment assignment was performed centrally using variable block sizes to ensure balance throughout trial recruitment and numbered drug packs were prepared and dispensed sequentially to randomised patients. The double-blind treatment was continued for 6 months in each subject.
  • LV mass was determined using cardiac MRI, performed in the supine position with the same 1.5 T scanner (Siemens Vision) using a phased array surface coil.
  • Prospectively gated cardiac cine images were acquired in 6 short axis and 3 long axis slices with the use of a segmented k-space pulse sequence (TR 8 ms; TE 5 ms; flip angle 10°; field of view 280-350 mm) with view sharing (11-19 frames.slice ⁇ 1 ). Each slice was obtained during a breath-hold of 15-19 heartbeats.
  • the short axis slices spanned the left ventricle from apex to base with a slice thickness of 8 mm and inter-slice gap of 2-6 mm.
  • the long axis slices were positioned at equal 60° intervals about the long axis of the LV.
  • Cardiac MRI provides accurate and reproducible estimates of LV mass and volume.
  • Table 7 shows baseline information on 30 patients with long-standing type 2 diabetes, no clinical evidence of coronary artery disease and abnormal diastolic function who participated in a 6-month randomized, double blind, placebo controlled study of chronic oral therapy with trientine dihydrochloride.
  • Trientine Placebo dihydrochloride N 15 15 Median age (years) 54 (range 43-64) 52 (range 33-69) % female 44% 56% Median duration of diabetes (years) 10 (1-24) 8 (1-21) Mean body mass index (kg/m 2 ) 32 (5) 34 (5) (SD) % hypertensive 64% 80% Mean % HbA 1c (SD) 9.3 (1.3) 9.3 (2.0)
  • LV masses were determined by tagged-molecular resonance imaging (MRI; see Bottini P B, et al., “Magnetic resonance imaging compared to echocardiography to assess left ventricular mass in the hypertensive patient,” Am. J. Hypertens 8: 221-228 (1995)) at baseline and following 6 months' trientine treatment. As expected, diabetics initially had significant LVH, consistent with previous reports. Struthers A D & Morris A D, “Screening for and treating left-ventricular abnormalities in diabetes mellitus: a new way of reducing cardiac deaths,” Lancet 359: 1430-1432 (2002).
  • MRI tagged-molecular resonance imaging
  • Results showed that Trientine treatment reverses LVH in type-2 diabetic humans.
  • MRI scans of the heart at baseline and 6-months showed a significant reduction in LV mass.
  • Mean LV mass in diabetics significantly decreased, by 5%, following 6 months' trientine treatment, whereas that in placebo-treated subjects increased by 3% ( FIG. 33 ); this highly significant effect remained after LV mass was indexed to body surface area, and occurred without change in systolic or diastolic blood pressure (Table 8).
  • trientine caused powerful regression in LV mass without altering blood pressure or urinary volume ( FIG. 32 ). No significant drug-related adverse events occurred during the 6 months' trientine therapy.
  • trientine administration for 6 months yielded improvements in the structure and function of the human heart.
  • This Example was carried out to assess the effect of chronic trientine administration on urinary metal excretion in diabetic and non-diabetic humans.
  • Trientine increased urinary Cu in both groups, but the excretion rate in diabetes was greater ( FIG. 30 ; P ⁇ 0.05).
  • trientine elicited similar urinary Cu responses in rats with T1DM and in humans with T2DM.
  • trientine In sun, chronic trientine administration increased urinary copper in both diabetic and nondiabetic groups, but the excretion rate in diabetes was greater. No corresponding increase in urinary Fe excretion was observed with trientine. Thus, trientine elicited similar urinary copper responses in rats with type 1 diabetes mellitus and in humans with type 2 diabetes mellitus.
  • This Example was carried out to determine the effect of oral trientine (triethylene tetramine dihydrochloride) administration on fecal output of metals in diabetic and non-diabetic humans. Methods were as follows.
  • T2DM type-2 diabetes mellitus
  • Total metal balance studies were performed in a residential metabolic unit
  • Total fecal outputs were collected daily for 12 days, freeze dried, and analyzed by ICP-MS for content of Cu, Fe, Zn, Ca, Mg, Mn, Cr, Mb and Se. Baseline measurements were taken during the first 6 d after which oral trientine or matched placebo were administered in a 2 ⁇ 2 randomized double-blind protocol and metal losses measured for a further 6 d.
  • Results were as follows. Mean daily fecal losses of Cu were not significantly different between subjects before and after administration of trientine or placebo, nor were Cu outputs different between diabetic and control subjects. The lack of effect of trientine on fecal Cu output was unexpected (see Table 11), and contrasts sharply with reports from Wilson's disease, in which trientine reportedly increased fecal Cu excretion.
  • This Example assessed the effect of the copper chelation efficacy of various concentrations of parenteral administration of trientine on anaesthetized diabetic and nondiabetic male Wistar rats through the measurement of copper in the urine.
  • Stock solutions of various intravenous formulations having concentrations of trientine hydrochloride were made up in 0.9% saline and was stored for four months at 4° C. without appreciable deterioriation in efficacy.
  • the concentrations of the stock formulations were: 0.67 mg/ml, 6.7 mg/ml, 67 mg/ml, and 670 mg/ml.
  • the formulation was then administered to the rats in doses of 0.1 mg/kg, 1 mg/kg, 10 mg/kg, and 100 mg/kg to the animals respectively.
  • the trachea was cannulated and the animal ventilated at 70-80 breaths.min ⁇ 1 with air supplemented with O2 (Pressure Controlled Ventilator, Kent Scientific, Connecticut, USA).
  • O2 Pressure Controlled Ventilator, Kent Scientific, Connecticut, USA.
  • the respiratory rate and end-tidal pressure (10-15 cmH2O) were adjusted to maintain end-tidal CO2 at 35-40 mmHg (SC-300 CO2 monitor, Pryon Corporation, Wisconsin, USA).
  • Body temperature was maintained at 37° C. throughout surgery and the experiment by a heating pad.
  • Estimated fluid loss was replaced with intravenous administration of 154 mmol.1 ⁇ 1 NaCl solution at a rate of 5 ml.kg ⁇ 1 .h ⁇ 1 .
  • MAP Mean arterial pressure
  • HR heart rate
  • HR heart rate
  • HR heart rate
  • HR heart rate
  • HR heart rate
  • core body temperature core body temperature
  • the trientine formulation or an equivalent volume of saline was intravenously administered hourly in doses of increasing strength from 0.1 mg/kg, 1.0 mg/kg, 10 mg/kg, and 100 mg/kg. Urine was collected throughout the experiment in 15 min aliquots.
  • Urine Urine was collected in pre-weighed 1.5 ml micro test tubes (eppendorf). After reweighing, the urine specimens were centrifuged and the supernatant diluted 25:1 with 0.02 m 69% Aristar grade HNO 3 . The sample was stored at 4° C. prior to GF-AAS analysis. If it was necessary to store a sample for a period in excess of 2 weeks, it was frozen and kept at ⁇ 20° C.
  • Serum Terminal blood samples were centrifuged and serum treated and stored as per urine until analysis.
  • trientine in the concentration ranges from between 0.1 mg/kg, 1 mg/kg, 10 mg/kg, and 100 mg/kg has no significant effect on blood pressure.
  • a trientine formulation is efficacious as a copper chelator when given intravenously and that trientine in saline remains active as a copper chelator after storage at 4° C. for 4 months.
  • This Example assessed the stability of a trientine formulation after being stored by its ability to chelate copper.
  • a standard 100 mM solution of Trientine HCl was made up in deionized (MilliQ) water.
  • One sample of the solution was stored in the dark at 4° C. and 21° C. in the dark and a third sample was stored at 21° C. in daylight.
  • the Ultraviolet-visible spectrum of the formulation was initially measured at day 0 and then at day 15. 20 ⁇ l aliquots of sample solutions were taken at day 15. For each aliquot 960 ⁇ l of 50 mM TRIS buffer and 20 ⁇ l aliquot of Copper Nitrate standard (100 mM—Orion Research Inc) were added. This was then measured over wavelengths 700-210 nm to determine the binding stability of the trientine formulations. See FIG. 26 that shows that there was no detectable change in the ability of the trientine formulation to chelate copper over this 15 day time period irrespective of storage conditions. Furthermore room light had no detectable detrimental effect on copper chelation and that trientine is stable as a chelator while in solution.

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Vascular Medicine (AREA)
  • Dermatology (AREA)
  • Emergency Medicine (AREA)
  • Toxicology (AREA)
  • Hospice & Palliative Care (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US10/525,345 2002-08-20 2003-08-20 Dosage forms and related therapies Abandoned US20060100278A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/573,211 US9339479B2 (en) 2002-08-20 2014-12-17 Dosage forms and related therapies
US15/096,570 US9993443B2 (en) 2002-08-20 2016-07-22 Dosage forms and related therapies
US15/966,433 US10543178B2 (en) 2002-08-20 2018-04-30 Dosage forms and related therapies
US16/719,404 US11419831B2 (en) 2002-08-20 2019-12-18 Dosage forms and related therapies

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
NZ520896 2002-08-20
NZ520895 2002-08-20
NZ52089702 2002-08-20
NZ520897 2002-08-20
NZ52089502 2002-08-20
NZ52089602 2002-08-20
NZ524795 2003-03-17
NZ52479403 2003-03-17
NZ524794 2003-03-17
NZ52479603 2003-03-17
NZ52479503 2003-03-17
NZ524796 2003-03-17
PCT/NZ2003/000184 WO2004017956A1 (fr) 2002-08-20 2003-08-20 Formes posologiques et traitements s'y rapportant

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/NZ2003/000184 A-371-Of-International WO2004017956A1 (fr) 2002-08-20 2003-08-20 Formes posologiques et traitements s'y rapportant

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/573,211 Continuation US9339479B2 (en) 2002-08-20 2014-12-17 Dosage forms and related therapies

Publications (1)

Publication Number Publication Date
US20060100278A1 true US20060100278A1 (en) 2006-05-11

Family

ID=31950924

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/525,345 Abandoned US20060100278A1 (en) 2002-08-20 2003-08-20 Dosage forms and related therapies
US14/573,211 Expired - Fee Related US9339479B2 (en) 2002-08-20 2014-12-17 Dosage forms and related therapies
US15/096,570 Expired - Fee Related US9993443B2 (en) 2002-08-20 2016-07-22 Dosage forms and related therapies
US15/966,433 Expired - Fee Related US10543178B2 (en) 2002-08-20 2018-04-30 Dosage forms and related therapies
US16/719,404 Expired - Lifetime US11419831B2 (en) 2002-08-20 2019-12-18 Dosage forms and related therapies

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/573,211 Expired - Fee Related US9339479B2 (en) 2002-08-20 2014-12-17 Dosage forms and related therapies
US15/096,570 Expired - Fee Related US9993443B2 (en) 2002-08-20 2016-07-22 Dosage forms and related therapies
US15/966,433 Expired - Fee Related US10543178B2 (en) 2002-08-20 2018-04-30 Dosage forms and related therapies
US16/719,404 Expired - Lifetime US11419831B2 (en) 2002-08-20 2019-12-18 Dosage forms and related therapies

Country Status (7)

Country Link
US (5) US20060100278A1 (fr)
EP (1) EP1539129A4 (fr)
JP (1) JP2006503014A (fr)
AU (1) AU2003258909B2 (fr)
CA (3) CA2875522A1 (fr)
MX (1) MXPA05002883A (fr)
WO (1) WO2004017956A1 (fr)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060009534A1 (en) * 2002-03-08 2006-01-12 Cooper Garth J Preventing and/or treating cardiovascular disease and/or associated heart failure
US20070054838A1 (en) * 2005-09-07 2007-03-08 Marcus Stark Method for improving diagnostic quality in echocardiography
US20070161120A1 (en) * 2006-01-10 2007-07-12 Viorica Lopez-Avila Ceruloplasmin Assay
US20080221722A1 (en) * 2007-03-08 2008-09-11 Popp Shane M Methods of interfacing nanomaterials for the monitoring and execution of pharmaceutical manufacturing processes
US20080312210A1 (en) * 2002-07-25 2008-12-18 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of a specific cyclic amine derivative or the pharmaceutically acceptable salts thereof for the treatment or prevention of heart failure
US20090004231A1 (en) * 2007-06-30 2009-01-01 Popp Shane M Pharmaceutical dosage forms fabricated with nanomaterials for quality monitoring
WO2009017802A1 (fr) * 2007-08-01 2009-02-05 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Acides gras nitrés pour le traitement du diabète
US20110098724A1 (en) * 2009-10-28 2011-04-28 Frank Cichocki Antimicrobial Coatings with Preferred Microstructure for Medical Devices
US20110268808A1 (en) * 2009-01-09 2011-11-03 Panacea Biotec Ltd. Dual-release pharmaceutical suspension
US8394992B2 (en) 2004-07-19 2013-03-12 Philera New Zealand Limited Synthesis of triethylenetetramines
US20160000834A1 (en) * 2013-02-28 2016-01-07 Ventrix, Inc. Methods and compositions for tissue therapy and analysis
US9289390B2 (en) 2011-08-12 2016-03-22 Boehringer Ingelheim Vetmedica Gmbh Taste masked pharmaceutical composition
WO2016126860A1 (fr) * 2015-02-03 2016-08-11 Kadmon Pharmaceuticals, Llc Formulations stables de trientine
US9585855B2 (en) 2008-06-19 2017-03-07 The University Of Utah Research Foundation Use of nitrated lipids for treatment of side effects of toxic medical therapies
US9663444B2 (en) 2009-10-02 2017-05-30 Complexa, Inc. Heteroatom containing substituted fatty acids
US9750725B2 (en) 2009-07-31 2017-09-05 University of Pittsburgh—of the Commonwealth System of Higher Education Fatty acids as anti-inflammatory agents
US9790167B2 (en) 2008-05-01 2017-10-17 Complexa, Inc. Vinyl substituted fatty acids
US10010532B2 (en) 2011-08-19 2018-07-03 The University Of Utah Research Foundation Combination therapy with nitrated lipids and inhibitors of the renin-angiotensin-aldosterone system
US20180296598A1 (en) * 2015-09-24 2018-10-18 Innolife Co., Ltd. Use of trientine to deliver copper to ischemic tissue
US10369125B2 (en) 2008-06-19 2019-08-06 The University Of Utah Research Foundation Method of treating renal system damage
CN110225905A (zh) * 2016-11-25 2019-09-10 Fng研究有限公司 用于保护皮肤免受重金属和甲醛影响的组合物
US10537541B2 (en) 2015-10-02 2020-01-21 Complexa Inc. Treatment of focal segmental glomerular sclerosis (FSGS) using therapeutically effective oral doses of 10-nitro-9(E)-octadec-9-enoic acid
WO2020264025A1 (fr) * 2019-06-28 2020-12-30 Passport Technologies, Inc. Timbre d'administration de perméant par l'intermédiaire d'un passage formé
US11077138B2 (en) * 2015-04-22 2021-08-03 Innolife Co., Ltd. Methods of tissue repair and regeneration
US11608342B2 (en) 2015-07-07 2023-03-21 H. Lundbeck A/S PDE9 inhibitors with imidazo triazinone backbone and imidazo pyrazinone backbone for treatment of peripheral diseases
US12006319B2 (en) 2018-05-25 2024-06-11 Cardurion Pharmaceuticals, Inc. Monohydrate and crystalline forms of 6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-3-tetrahydropyran-4-yl-7H-imidazo[1,5-a]pyrazin-8-one

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8198334B2 (en) * 1997-10-27 2012-06-12 Pathologica Llc Methods for modulating macrophage proliferation in ocular disease using polyamine analogs
EP1115389B1 (fr) 1998-09-25 2014-03-12 PhilERA New Zealand Limited Fructosamine-oxydase: antagonistes et inhibiteurs
CA2875522A1 (fr) 2002-08-20 2004-03-04 Philera New Zealand Limited Formes posologiques et traitements s'y rapportant
WO2006068516A1 (fr) * 2004-12-20 2006-06-29 Protemix Corporation Limited Dispositifs medicaux implantables revetus de composes chelateurs du cuivre ou contenant ces composes
TW201609145A (zh) * 2013-12-25 2016-03-16 參天製藥股份有限公司 注射劑及形成緩釋(depot)之方法
CN107133397B (zh) * 2017-04-27 2018-10-19 山东大学 一种基于ale法对生物瓣膜进行双向流固耦合分析的方法
CA3069921C (fr) * 2017-07-19 2022-01-18 Primus Pharmaceuticals, Inc. Methodes de traitement de troubles microvasculaires auto-immuns
US11471497B1 (en) 2019-03-13 2022-10-18 David Gordon Bermudes Copper chelation therapeutics
CN114581594A (zh) * 2020-12-18 2022-06-03 完美世界(北京)软件科技发展有限公司 次表面散射效果的渲染方法及装置、设备、介质
EP4301350A1 (fr) * 2021-03-05 2024-01-10 Philera New Zealand Ltd. Traitement de troubles liés au cuivre
US20240130985A1 (en) * 2022-10-19 2024-04-25 Genexa Inc. Solid pharmaceutical tablet

Citations (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791988A (en) * 1972-03-23 1974-02-12 Hoffmann La Roche Diagnostic test for glucose
US4323558A (en) * 1979-09-10 1982-04-06 Nelson Research & Development Co. Topical trien containing pharmaceutical compositions and methods of use
US4374829A (en) * 1978-12-11 1983-02-22 Merck & Co., Inc. Aminoacid derivatives as antihypertensives
US4410541A (en) * 1981-03-02 1983-10-18 Nihon Nosan Kogyo K.K. Composite for improving lipid metabolism
US4758583A (en) * 1984-03-19 1988-07-19 The Rockefeller University Method and agents for inhibiting protein aging
US4866090A (en) * 1988-01-07 1989-09-12 Merck & Co., Inc. Novel HMG-CoA reductase inhibitors
US4952568A (en) * 1988-05-19 1990-08-28 Sanwa Kagaku Kenkyusho Co., Ltd. Remedies and preventives for diabetic diseases
US5077313A (en) * 1988-11-25 1991-12-31 Gert Lubec Process for inhibiting pathological collagen cross-linking in diabetes patients
US5128360A (en) * 1984-03-19 1992-07-07 The Rockefeller University Method and agents for inhibiting protein aging
US5246970A (en) * 1991-12-16 1993-09-21 Washington University Method of inhibiting nitric oxide formation
US5366505A (en) * 1991-12-06 1994-11-22 North Shore University Hospital Research Corporation Method of reducing medical device related infections
US5387109A (en) * 1992-06-05 1995-02-07 Nakano Vinegar Co., Ltd. Fructosylamine deglycase and a method of producing it
US5420120A (en) * 1993-12-17 1995-05-30 Alcon Laboratories, Inc. Anti-inflammatory glucocorticoid compounds for topical ophthalmic use
US5811446A (en) * 1997-04-18 1998-09-22 Cytos Pharmaceuticals Llc Prophylactic and therapeutic methods for ocular degenerative diseases and inflammations and histidine compositions therefor
US5824049A (en) * 1995-06-07 1998-10-20 Med Institute, Inc. Coated implantable medical device
US5852009A (en) * 1984-03-19 1998-12-22 The Rockefeller University Compositions, including pharmaceutical compositions, for inhibiting the advanced glycosylation of proteins, and therapeutic methods based thereon
US5854271A (en) * 1994-05-02 1998-12-29 Cytos Pharmaceuticals, L.L.C. Effective method for the amelioration and prevention of tissue and cellular damage
US5906996A (en) * 1996-08-21 1999-05-25 Murphy; Michael A. Tetramine treatment of neurological disorders
US5972985A (en) * 1997-11-03 1999-10-26 Cytos Pharmaceuticals, Llc Histidine containing nutriceutical compositions
US5980914A (en) * 1997-08-22 1999-11-09 P.N. Gerolymatos S.A. Clioquinol for the treatment of Parkinson's disease
US6147070A (en) * 1998-06-05 2000-11-14 Facchini; Francesco Methods and compositions for controlling iron stores to treat and cure disease states
US6309380B1 (en) * 1999-01-27 2001-10-30 Marian L. Larson Drug delivery via conformal film
US6348465B1 (en) * 1998-09-25 2002-02-19 Protemix Corporation Limited Fructosamine oxidase: antagonists and inhibitors
US20020031775A1 (en) * 1999-12-21 2002-03-14 Erikson Glen H. Parallel or antiparallel, homologous or complementary binding of nucleic acids or analogues thereof to form duplex, triplex or quadruplex complexes
US20030013772A1 (en) * 2000-02-23 2003-01-16 Murphy Michael A. Composition, synthesis and therapeutic applications of polyamines
US20030055003A1 (en) * 2001-07-19 2003-03-20 David Bar-Or Use of copper chelators to inhibit the inactivation of protein C
US20030055113A1 (en) * 2000-12-01 2003-03-20 Xuefeng Wang Copper chelators for treating ocular inflammation
US6576672B1 (en) * 1998-08-21 2003-06-10 Michael A. Murphy Polyamine treatment of neurological disorders
US20030139312A1 (en) * 2000-05-11 2003-07-24 Caswell Debra Sue Highly concentrated fabric softener compositions and articles containing such compositions
US20030166561A1 (en) * 1999-06-18 2003-09-04 Cooper Garth J. S. Peptide
US20030186946A1 (en) * 2002-01-29 2003-10-02 Cooper Garth James Smith Suppression of cytotoxic protein conformers
US20030203973A1 (en) * 2002-03-08 2003-10-30 Cooper Garth J. S. Preventing and/or treating cardiovascular disease and/or associated heart failure
US20030232799A1 (en) * 2000-12-01 2003-12-18 Xuefeng Wang Copper chelators for treating ocular Inflammation
US20040019087A1 (en) * 2002-07-23 2004-01-29 Ternansky Robert J. Thiomolybdate analogues and uses thereof
US20040023854A1 (en) * 2002-01-18 2004-02-05 Cooper Garth J.S. Adiponectin and uses thereof
US20040038861A1 (en) * 2001-11-26 2004-02-26 Cooper Garth J. S. Methods and compositions for normalizing lipid levels in mammalian tissues
US20040076603A1 (en) * 2002-01-25 2004-04-22 Tony Peled Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US20040142393A1 (en) * 2002-08-01 2004-07-22 Cooper Garth James Smith Methods of use of compounds with preptin function
US6821954B2 (en) * 1997-09-18 2004-11-23 Auckland Uniservices Limited Compounds and uses thereof in treating bone disorders
US20040259945A1 (en) * 2002-07-23 2004-12-23 The Regents Of The University Of Michigan Tetrapropylammonium tetrathiomolybdate and related compounds for anti-angiogenic therapies
US20050002876A1 (en) * 2003-05-07 2005-01-06 Yukl Richard L. Oral care methods and products
US20050009760A1 (en) * 2003-07-11 2005-01-13 Suming Wang Methods and compositions for treatment of ataxia-telangeictasia
US20050031651A1 (en) * 2002-12-24 2005-02-10 Francine Gervais Therapeutic formulations for the treatment of beta-amyloid related diseases
US6855511B2 (en) * 1998-09-25 2005-02-15 Protemix Corporation Limited Fructosamine oxidase assay: methods and materials
US20050047998A1 (en) * 2001-03-30 2005-03-03 Cooper Garth J.S. Phosphoprotein target for insulin and its antagonists
US20050074756A1 (en) * 2002-03-01 2005-04-07 Cooper Garth James Smith FALP proteins
US20050085555A1 (en) * 1997-08-21 2005-04-21 Murphy Michael A. Composition, synthesis and therapeutic applications of polyamines
US20050159364A1 (en) * 2003-12-19 2005-07-21 Cooper Garth J. Copper antagonist compounds

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL105793B1 (pl) 1977-11-24 1979-10-31 Sposob otrzymywania soli kwasow dwukarboksylowych i poliamin
US4371374A (en) 1980-11-17 1983-02-01 The Rockefeller University Monitoring metabolic control in diabetic patients by measuring glycosylated amino acids and peptides in urine
DE3217071A1 (de) 1982-05-06 1983-11-10 Rüdiger Dr. 6365 Rosbach Gröning Arzneimittel fuer die perorale applikation mit verbesserter biologischer verfuegbarkeit der wirkstoffe
DE3750369T2 (de) 1986-03-21 1995-05-24 Eurasiam Lab Arzneimittelzusammensetzungen.
CH667590A5 (it) 1986-07-24 1988-10-31 Inpharzam Int Sa Composizione farmaceutica effervescente idrosolubile contenente n-acetil-cisteina. .
CH666814A5 (it) 1986-07-24 1988-08-31 Inpharzam Int Sa Composizione farmaceutica idrosolubile contenente n-acetil-cisteina.
EP0331014A3 (fr) 1988-03-02 1991-10-23 THERA - Patent Verwaltungs-GmbH L'utilisation des inhibiteurs de l'enzyme de conversion pour la prophylaxie du diabète
CA2026686A1 (fr) 1989-10-30 1991-05-01 Werner Tschollar Methode de prevention de l'apparition du diabete de type 11 par l'emploi d'un inhibiteur de l'enzyme de conversion
US5447728A (en) 1992-06-15 1995-09-05 Emisphere Technologies, Inc. Desferrioxamine oral delivery system
JPH07118148A (ja) 1993-10-26 1995-05-09 Tsumura & Co 肝癌予防剤
WO1995017900A1 (fr) 1993-12-30 1995-07-06 Arcturus Pharmaceutical Corporation Methodes de traitement de troubles du systeme nerveux central ou de l'oeil impliquant des voies d'oxydation pathogene
AU4134996A (en) 1994-10-25 1996-05-15 Washington University Method of inhibiting nitric oxide formation
US6004958A (en) * 1997-02-05 1999-12-21 Fox Chase Cancer Center Compounds and methods for therapeutic intervention in preventing diabetic complications and procedures for assessing a diabetic's risk of developing complications and determining the efficacy of therapeutic intervention
JP2001514661A (ja) 1997-03-11 2001-09-11 ザ ジェネラル ホスピタル コーポレイション アルツハイマー病の処置における使用のための薬剤の同定
JP2002511387A (ja) 1998-02-04 2002-04-16 ユーロ−セルティック エス. ア. 置換セミカルバジドおよびその使用
US6323218B1 (en) 1998-03-11 2001-11-27 The General Hospital Corporation Agents for use in the treatment of Alzheimer's disease
SI1107795T1 (en) * 1998-09-04 2003-06-30 The Regents Of The University Of Michigan Thiomolybdate associated with at least one carbohydrate, and its use for preventing or treating diseases characterized by aberrant vascularization, such as cancer, wet type macular degeneration, rheumatoid arthritis
JP2000204037A (ja) 1999-01-12 2000-07-25 Tsumura & Co 筋萎縮性側索硬化症治療薬
CN1296384C (zh) 1999-06-18 2007-01-24 普罗特米克斯发现有限公司 具有preptin功能的肽
US20030050434A1 (en) 1999-06-18 2003-03-13 Cooper Garth J.S. Peptide
JPWO2002073722A1 (ja) 2001-03-08 2004-07-08 ソニー株式会社 ガス拡散性電極体及びその製造方法、並びに電気化学デバイス
CA2471833A1 (fr) 2001-11-26 2003-06-05 Protemix Corporation Limited Procedes et compositions pour normaliser les niveaux lipidiques dans les tissus mammaliens
US7144706B2 (en) 2002-01-03 2006-12-05 Duke University Methods of modulating localization and physiological function of IP3 receptors
WO2003075910A1 (fr) * 2002-03-08 2003-09-18 Protemix Corporation Limited Prevention et/ou traitement d'une maladie vasculaire, d'une cardiomyopathie et/ou d'une defaillance cardiaque associee
EP1494658B1 (fr) 2002-04-03 2006-01-04 Puleva Biotech, S.A. Produits phenoliques naturels et leurs derives permettant d'obtenir une protection contre les maladies neurodegeneratives
AU2003222533A1 (en) 2002-04-29 2003-11-17 Protemix Corporation Limited Proteins with deglycating activities and methods of using same
WO2003099223A2 (fr) * 2002-05-24 2003-12-04 The Regents Of The University Of Michigan Traitement reduisant le taux de cuivre pour des maladies inflammatoires et fibreuses
CA2494308A1 (fr) 2002-08-01 2004-02-12 Protemix Corporation Limited Methodes d'utilisation de composes avec une fonction preptine
CA2494305A1 (fr) 2002-08-01 2004-02-12 Auckland Uniservices Limited Procedes d'utilisation de la preptine
CA2875522A1 (fr) 2002-08-20 2004-03-04 Philera New Zealand Limited Formes posologiques et traitements s'y rapportant
WO2004017957A1 (fr) 2002-08-20 2004-03-04 Protemix Corporation Limited Prevention et/ou traitement de maladies cardio-vasculaires et/ou insuffisance cardiaque associee
AU2003285686A1 (en) 2002-12-23 2004-07-14 Protemix Corporation Limited Glycoisoforms of adiponectin and uses thereof
WO2004083215A2 (fr) 2003-03-21 2004-09-30 Palumed Sa Derives polycycliques azotes utilises comme chelateurs d'ions metalliques et leurs applications
US20070037848A1 (en) 2003-04-03 2007-02-15 Masters Colin L Treatment of neurological conditions
US6927206B2 (en) 2003-06-06 2005-08-09 Procyte Corporation Compositions and methods for treatment of rosacea
EP1687330A4 (fr) 2003-10-28 2007-03-14 Protemix Discovery Ltd Peptides avec action contre l'obesite et autre utilisations apparentees
CA3050047A1 (fr) 2004-07-19 2006-03-16 Philera New Zealand Limited Synthese de triethylenetetramines
WO2006062422A1 (fr) 2004-12-10 2006-06-15 Protemix Corporation Limited Glyponectine (adiponectine glycosylee) pour le traitement de maladies et d'etats pathologiques
CN112334731B (zh) 2018-06-27 2022-09-02 东京毅力科创株式会社 膜厚测定装置及校正方法

Patent Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791988A (en) * 1972-03-23 1974-02-12 Hoffmann La Roche Diagnostic test for glucose
US4374829A (en) * 1978-12-11 1983-02-22 Merck & Co., Inc. Aminoacid derivatives as antihypertensives
US4323558A (en) * 1979-09-10 1982-04-06 Nelson Research & Development Co. Topical trien containing pharmaceutical compositions and methods of use
US4410541A (en) * 1981-03-02 1983-10-18 Nihon Nosan Kogyo K.K. Composite for improving lipid metabolism
US5128360A (en) * 1984-03-19 1992-07-07 The Rockefeller University Method and agents for inhibiting protein aging
US4758583A (en) * 1984-03-19 1988-07-19 The Rockefeller University Method and agents for inhibiting protein aging
US5852009A (en) * 1984-03-19 1998-12-22 The Rockefeller University Compositions, including pharmaceutical compositions, for inhibiting the advanced glycosylation of proteins, and therapeutic methods based thereon
US4866090A (en) * 1988-01-07 1989-09-12 Merck & Co., Inc. Novel HMG-CoA reductase inhibitors
US4952568A (en) * 1988-05-19 1990-08-28 Sanwa Kagaku Kenkyusho Co., Ltd. Remedies and preventives for diabetic diseases
US5077313A (en) * 1988-11-25 1991-12-31 Gert Lubec Process for inhibiting pathological collagen cross-linking in diabetes patients
US5366505A (en) * 1991-12-06 1994-11-22 North Shore University Hospital Research Corporation Method of reducing medical device related infections
US5246970A (en) * 1991-12-16 1993-09-21 Washington University Method of inhibiting nitric oxide formation
US5387109A (en) * 1992-06-05 1995-02-07 Nakano Vinegar Co., Ltd. Fructosylamine deglycase and a method of producing it
US5420120A (en) * 1993-12-17 1995-05-30 Alcon Laboratories, Inc. Anti-inflammatory glucocorticoid compounds for topical ophthalmic use
US5854271A (en) * 1994-05-02 1998-12-29 Cytos Pharmaceuticals, L.L.C. Effective method for the amelioration and prevention of tissue and cellular damage
US5824049A (en) * 1995-06-07 1998-10-20 Med Institute, Inc. Coated implantable medical device
US5906996A (en) * 1996-08-21 1999-05-25 Murphy; Michael A. Tetramine treatment of neurological disorders
US5811446A (en) * 1997-04-18 1998-09-22 Cytos Pharmaceuticals Llc Prophylactic and therapeutic methods for ocular degenerative diseases and inflammations and histidine compositions therefor
US20050085555A1 (en) * 1997-08-21 2005-04-21 Murphy Michael A. Composition, synthesis and therapeutic applications of polyamines
US5980914A (en) * 1997-08-22 1999-11-09 P.N. Gerolymatos S.A. Clioquinol for the treatment of Parkinson's disease
US6821954B2 (en) * 1997-09-18 2004-11-23 Auckland Uniservices Limited Compounds and uses thereof in treating bone disorders
US5972985A (en) * 1997-11-03 1999-10-26 Cytos Pharmaceuticals, Llc Histidine containing nutriceutical compositions
US6329414B1 (en) * 1997-11-03 2001-12-11 Cytos Pharmaceuticals Llc Histidine containing nutriceutical
US6147070A (en) * 1998-06-05 2000-11-14 Facchini; Francesco Methods and compositions for controlling iron stores to treat and cure disease states
US6576672B1 (en) * 1998-08-21 2003-06-10 Michael A. Murphy Polyamine treatment of neurological disorders
US6610693B2 (en) * 1998-09-19 2003-08-26 Protemix Corporation Limited Fructosamine oxidase: antagonists and inhibitors
US6348465B1 (en) * 1998-09-25 2002-02-19 Protemix Corporation Limited Fructosamine oxidase: antagonists and inhibitors
US20050159489A1 (en) * 1998-09-25 2005-07-21 Baker John R. Treatment of diabetes with copper binding compounds
US6897243B2 (en) * 1998-09-25 2005-05-24 Protemix Corporation Limited Fructosamine oxidase: antagonists and inhibitors
US20030045506A1 (en) * 1998-09-25 2003-03-06 Baker John Richard Fructosamine oxidase: antagonists and inhibitors
US6855511B2 (en) * 1998-09-25 2005-02-15 Protemix Corporation Limited Fructosamine oxidase assay: methods and materials
US6309380B1 (en) * 1999-01-27 2001-10-30 Marian L. Larson Drug delivery via conformal film
US20030166561A1 (en) * 1999-06-18 2003-09-04 Cooper Garth J. S. Peptide
US20020031775A1 (en) * 1999-12-21 2002-03-14 Erikson Glen H. Parallel or antiparallel, homologous or complementary binding of nucleic acids or analogues thereof to form duplex, triplex or quadruplex complexes
US20030013772A1 (en) * 2000-02-23 2003-01-16 Murphy Michael A. Composition, synthesis and therapeutic applications of polyamines
US20030139312A1 (en) * 2000-05-11 2003-07-24 Caswell Debra Sue Highly concentrated fabric softener compositions and articles containing such compositions
US20030055113A1 (en) * 2000-12-01 2003-03-20 Xuefeng Wang Copper chelators for treating ocular inflammation
US20030232799A1 (en) * 2000-12-01 2003-12-18 Xuefeng Wang Copper chelators for treating ocular Inflammation
US20050047998A1 (en) * 2001-03-30 2005-03-03 Cooper Garth J.S. Phosphoprotein target for insulin and its antagonists
US6884575B2 (en) * 2001-03-30 2005-04-26 Protemix Corporation Limited Phosphoprotein target for insulin and its antagonists
US20030055003A1 (en) * 2001-07-19 2003-03-20 David Bar-Or Use of copper chelators to inhibit the inactivation of protein C
US20040038861A1 (en) * 2001-11-26 2004-02-26 Cooper Garth J. S. Methods and compositions for normalizing lipid levels in mammalian tissues
US20040023854A1 (en) * 2002-01-18 2004-02-05 Cooper Garth J.S. Adiponectin and uses thereof
US20040076603A1 (en) * 2002-01-25 2004-04-22 Tony Peled Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US20030186946A1 (en) * 2002-01-29 2003-10-02 Cooper Garth James Smith Suppression of cytotoxic protein conformers
US20050074756A1 (en) * 2002-03-01 2005-04-07 Cooper Garth James Smith FALP proteins
US6951890B2 (en) * 2002-03-08 2005-10-04 Protemix Corporation Limited Preventing and/or treating cardiovascular disease and/or associated heart failure
US20030203973A1 (en) * 2002-03-08 2003-10-30 Cooper Garth J. S. Preventing and/or treating cardiovascular disease and/or associated heart failure
US20040259945A1 (en) * 2002-07-23 2004-12-23 The Regents Of The University Of Michigan Tetrapropylammonium tetrathiomolybdate and related compounds for anti-angiogenic therapies
US20040019087A1 (en) * 2002-07-23 2004-01-29 Ternansky Robert J. Thiomolybdate analogues and uses thereof
US20040142393A1 (en) * 2002-08-01 2004-07-22 Cooper Garth James Smith Methods of use of compounds with preptin function
US20050031651A1 (en) * 2002-12-24 2005-02-10 Francine Gervais Therapeutic formulations for the treatment of beta-amyloid related diseases
US20050002876A1 (en) * 2003-05-07 2005-01-06 Yukl Richard L. Oral care methods and products
US20050009760A1 (en) * 2003-07-11 2005-01-13 Suming Wang Methods and compositions for treatment of ataxia-telangeictasia
US20050159364A1 (en) * 2003-12-19 2005-07-21 Cooper Garth J. Copper antagonist compounds

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060009534A1 (en) * 2002-03-08 2006-01-12 Cooper Garth J Preventing and/or treating cardiovascular disease and/or associated heart failure
US8034799B2 (en) 2002-03-08 2011-10-11 Philera New Zealand Limited Preventing and/or treating cardiovascular disease and/or associated heart failure
US20080312210A1 (en) * 2002-07-25 2008-12-18 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of a specific cyclic amine derivative or the pharmaceutically acceptable salts thereof for the treatment or prevention of heart failure
US8524701B2 (en) 2002-07-25 2013-09-03 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of a specific cyclic amine derivative or the pharmaceutically acceptable salts thereof for the treatment or prevention of heart failure
US11795150B2 (en) 2004-07-19 2023-10-24 Philera New Zealand Limited Synthesis of triethylenetetramines
US9556123B2 (en) 2004-07-19 2017-01-31 Philera New Zealand Limited Synthesis of triethylenetetramines
US8912362B2 (en) 2004-07-19 2014-12-16 Philera New Zealand Limited Synthesis of triethylenetetramines
US8394992B2 (en) 2004-07-19 2013-03-12 Philera New Zealand Limited Synthesis of triethylenetetramines
US20070054838A1 (en) * 2005-09-07 2007-03-08 Marcus Stark Method for improving diagnostic quality in echocardiography
US20070161120A1 (en) * 2006-01-10 2007-07-12 Viorica Lopez-Avila Ceruloplasmin Assay
US7680553B2 (en) 2007-03-08 2010-03-16 Smp Logic Systems Llc Methods of interfacing nanomaterials for the monitoring and execution of pharmaceutical manufacturing processes
US20100217425A1 (en) * 2007-03-08 2010-08-26 Popp Shane M Manufacturing execution system (MES) and methods of monitoring glycol manufacturing processes utilizing functional nanomaterials
US20080221722A1 (en) * 2007-03-08 2008-09-11 Popp Shane M Methods of interfacing nanomaterials for the monitoring and execution of pharmaceutical manufacturing processes
US20090004231A1 (en) * 2007-06-30 2009-01-01 Popp Shane M Pharmaceutical dosage forms fabricated with nanomaterials for quality monitoring
US10060860B2 (en) 2007-06-30 2018-08-28 Smp Logic Systems Pharmaceutical dosage forms fabricated with nanomaterials
US10576051B2 (en) 2007-08-01 2020-03-03 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
US10258589B2 (en) 2007-08-01 2019-04-16 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
US10869850B2 (en) 2007-08-01 2020-12-22 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
WO2009017802A1 (fr) * 2007-08-01 2009-02-05 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Acides gras nitrés pour le traitement du diabète
US9186408B2 (en) 2007-08-01 2015-11-17 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
US20100216884A1 (en) * 2007-08-01 2010-08-26 University of Pittsburg - of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type ii diabetes
US9700534B2 (en) 2007-08-01 2017-07-11 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
US8735449B2 (en) 2007-08-01 2014-05-27 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
US8324277B2 (en) 2007-08-01 2012-12-04 University of Pittsburgh—of the Commonwealth System of Higher Education Nitrated-fatty acids modulation of type II diabetes
US9790167B2 (en) 2008-05-01 2017-10-17 Complexa, Inc. Vinyl substituted fatty acids
US9585855B2 (en) 2008-06-19 2017-03-07 The University Of Utah Research Foundation Use of nitrated lipids for treatment of side effects of toxic medical therapies
US10369125B2 (en) 2008-06-19 2019-08-06 The University Of Utah Research Foundation Method of treating renal system damage
US10568857B2 (en) 2008-06-19 2020-02-25 The University Of Utah Research Foundation Method of treating renal system damage
US20110268808A1 (en) * 2009-01-09 2011-11-03 Panacea Biotec Ltd. Dual-release pharmaceutical suspension
US9750725B2 (en) 2009-07-31 2017-09-05 University of Pittsburgh—of the Commonwealth System of Higher Education Fatty acids as anti-inflammatory agents
US11723897B2 (en) 2009-07-31 2023-08-15 University of Pittsburgh—of the Commonwealth System of Higher Education Fatty acids as anti-inflammatory agents
US10835518B2 (en) 2009-07-31 2020-11-17 University of Pittsburgh—of the Commonwealth System of Higher Education Fatty acids as anti-inflammatory agents
US10213417B2 (en) 2009-07-31 2019-02-26 University of Pittsburgh—of the Commonwealth System of Higher Education Fatty acids as anti-inflammatory agents
US9663444B2 (en) 2009-10-02 2017-05-30 Complexa, Inc. Heteroatom containing substituted fatty acids
US20110098724A1 (en) * 2009-10-28 2011-04-28 Frank Cichocki Antimicrobial Coatings with Preferred Microstructure for Medical Devices
US8808724B2 (en) * 2009-10-28 2014-08-19 Ethicon, Inc. Antimicrobial coatings with preferred microstructure for medical devices
US9289390B2 (en) 2011-08-12 2016-03-22 Boehringer Ingelheim Vetmedica Gmbh Taste masked pharmaceutical composition
US10709690B2 (en) 2011-08-19 2020-07-14 The University Of Utah Research Foundation Combination therapy with nitrated lipids and inhibitors of the renin-angiotensin-aldosterone system
US10010532B2 (en) 2011-08-19 2018-07-03 The University Of Utah Research Foundation Combination therapy with nitrated lipids and inhibitors of the renin-angiotensin-aldosterone system
US20160000834A1 (en) * 2013-02-28 2016-01-07 Ventrix, Inc. Methods and compositions for tissue therapy and analysis
US10874624B2 (en) 2015-02-03 2020-12-29 Kadmon Pharmaceuticals, Llc Stable trientine formulations
WO2016126860A1 (fr) * 2015-02-03 2016-08-11 Kadmon Pharmaceuticals, Llc Formulations stables de trientine
JP2018505215A (ja) * 2015-02-03 2018-02-22 カドモン ファーマシューティカルズ,リミティド ライアビリティ カンパニー 安定トリエンチン製剤
CN107530311A (zh) * 2015-02-03 2018-01-02 卡德门制药有限公司 稳定的曲恩汀制剂
US11077138B2 (en) * 2015-04-22 2021-08-03 Innolife Co., Ltd. Methods of tissue repair and regeneration
US11608342B2 (en) 2015-07-07 2023-03-21 H. Lundbeck A/S PDE9 inhibitors with imidazo triazinone backbone and imidazo pyrazinone backbone for treatment of peripheral diseases
US11033579B2 (en) 2015-09-24 2021-06-15 Innolife Co., Ltd. Use of trientine to deliver copper to ischemic tissue
US20180296598A1 (en) * 2015-09-24 2018-10-18 Innolife Co., Ltd. Use of trientine to deliver copper to ischemic tissue
AU2016328156B2 (en) * 2015-09-24 2022-03-17 Innolife Co., Ltd. Use of trientine to deliver copper to ischemic tissue
CN108697669A (zh) * 2015-09-24 2018-10-23 北京因诺瑞康生物医药科技有限公司 曲恩汀将铜递送到缺血组织的用途
US10537541B2 (en) 2015-10-02 2020-01-21 Complexa Inc. Treatment of focal segmental glomerular sclerosis (FSGS) using therapeutically effective oral doses of 10-nitro-9(E)-octadec-9-enoic acid
CN110225905A (zh) * 2016-11-25 2019-09-10 Fng研究有限公司 用于保护皮肤免受重金属和甲醛影响的组合物
US12006319B2 (en) 2018-05-25 2024-06-11 Cardurion Pharmaceuticals, Inc. Monohydrate and crystalline forms of 6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-3-tetrahydropyran-4-yl-7H-imidazo[1,5-a]pyrazin-8-one
WO2020264025A1 (fr) * 2019-06-28 2020-12-30 Passport Technologies, Inc. Timbre d'administration de perméant par l'intermédiaire d'un passage formé

Also Published As

Publication number Publication date
US9339479B2 (en) 2016-05-17
US20200276135A1 (en) 2020-09-03
CA2496411A1 (fr) 2004-03-04
CA2875522A1 (fr) 2004-03-04
EP1539129A4 (fr) 2006-03-08
EP1539129A1 (fr) 2005-06-15
US20150164826A1 (en) 2015-06-18
AU2003258909B2 (en) 2010-07-08
US9993443B2 (en) 2018-06-12
CA3011023A1 (fr) 2004-03-04
WO2004017956A1 (fr) 2004-03-04
JP2006503014A (ja) 2006-01-26
US20190000778A1 (en) 2019-01-03
MXPA05002883A (es) 2005-10-05
US20160324804A1 (en) 2016-11-10
US10543178B2 (en) 2020-01-28
US11419831B2 (en) 2022-08-23
AU2003258909A1 (en) 2004-03-11

Similar Documents

Publication Publication Date Title
US11419831B2 (en) Dosage forms and related therapies
US20050159364A1 (en) Copper antagonist compounds
JP6515121B2 (ja) 医薬組成物、治療方法及びその使用
JP5611275B2 (ja) 真性糖尿病の治療のためのロフルミラスト
EP1303304B1 (fr) Combinaisons pharmaceutiques pour traiter et prevenir diabetes mellitus
JP6066144B2 (ja) 併用医薬
ES2241305T3 (es) Combinaciones contra la diabetes que contienen gliburide, troglitazona (y una biguanida).
JPH10513455A (ja) 緑内障治療用のデプレニル化合物
WO2020072814A1 (fr) Composés biphényl-sulfonamide destinés au traitement de maladies du collagène de type iv
US20140100282A1 (en) Intranasal administration of pharmaceutical agents for treatment of neurological diseases
WO2002009701A1 (fr) Utilisation d'agonistes, d'antagonistes et d'amplificateurs allosteriques selectifs du recepteur d'adenosine a1 afin de manipuler l'angiogenese
WO2014008374A2 (fr) Thérapies combinées comprenant des sels de metformine et des agents antihyperglycémie ou des agents antihyperlipidémie
US20090093542A1 (en) Copper antagonist compositions
NZ539695A (en) Dosage forms and related therapies
JP2002542295A (ja) 哺乳動物において、インスリン感受性およびグルコース代謝を改善する組成物および方法
WO2007011972A2 (fr) Profil pharmocinetique ameliore d'agonistes inverses beta-adrenergiques pour le traitement de maladies des voies aeriennes pulmonaires
US10292949B2 (en) Pharmaceutical composition comprising rimeporide for treating diseases associated with insulin resistance and β-cell dysfunction
WO2006104398A1 (fr) Compositions antagonistes du cuivre
WO2006104402A1 (fr) Compositions d'antagonistes du cuivre
DE60305592T2 (de) Liposomenhaltiges Mittel zur Behandlung Gefässerkrankungen
EP1746993A1 (fr) Compositions a base de metformine a liberation regulee

Legal Events

Date Code Title Description
AS Assignment

Owner name: PROTEMIX CORPORATION LIMITED, NEW ZEALAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COOPER, GARTH JAMES SMITH;BAKER, JOHN RICHARD;BEELEY, NIGEL ROBERT ARNOLD;REEL/FRAME:016645/0605;SIGNING DATES FROM 20050318 TO 20050811

AS Assignment

Owner name: PHILERA NEW ZEALAND LIMITED, NEW ZEALAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PROTEMIX CORPORATION LIMITED;REEL/FRAME:026230/0428

Effective date: 20101207

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION