WO2002096347A2 - Metering and packaging of controlled release medication - Google Patents

Metering and packaging of controlled release medication Download PDF

Info

Publication number
WO2002096347A2
WO2002096347A2 PCT/US2002/016185 US0216185W WO02096347A2 WO 2002096347 A2 WO2002096347 A2 WO 2002096347A2 US 0216185 W US0216185 W US 0216185W WO 02096347 A2 WO02096347 A2 WO 02096347A2
Authority
WO
WIPO (PCT)
Prior art keywords
combination
delivery package
pharmaceutical delivery
plus
pharmaceutical
Prior art date
Application number
PCT/US2002/016185
Other languages
French (fr)
Other versions
WO2002096347A3 (en
Inventor
Andrew L. Abrams
Anand V. Gumaste
Original Assignee
Microdose Technologies, Inc.
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 Microdose Technologies, Inc. filed Critical Microdose Technologies, Inc.
Priority to US10/479,438 priority Critical patent/US20040156903A1/en
Priority to CA002448997A priority patent/CA2448997A1/en
Priority to EP02731902A priority patent/EP1408907A4/en
Priority to NZ529696A priority patent/NZ529696A/en
Priority to BRPI0209720A priority patent/BRPI0209720A2/en
Priority to JP2002592860A priority patent/JP2005514966A/en
Priority to AU2002303840A priority patent/AU2002303840A1/en
Publication of WO2002096347A2 publication Critical patent/WO2002096347A2/en
Publication of WO2002096347A3 publication Critical patent/WO2002096347A3/en
Priority to US11/549,492 priority patent/US20070087048A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61JCONTAINERS SPECIALLY ADAPTED FOR MEDICAL OR PHARMACEUTICAL PURPOSES; DEVICES OR METHODS SPECIALLY ADAPTED FOR BRINGING PHARMACEUTICAL PRODUCTS INTO PARTICULAR PHYSICAL OR ADMINISTERING FORMS; DEVICES FOR ADMINISTERING FOOD OR MEDICINES ORALLY; BABY COMFORTERS; DEVICES FOR RECEIVING SPITTLE
    • A61J3/00Devices or methods specially adapted for bringing pharmaceutical products into particular physical or administering forms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • A61K9/209Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat containing drug in at least two layers or in the core and in at least one outer layer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4808Preparations in capsules, e.g. of gelatin, of chocolate characterised by the form of the capsule or the structure of the filling; Capsules containing small tablets; Capsules with outer layer for immediate drug release

Definitions

  • the present invention relates to the metering and packaging of precise quantities of pharmaceuticals and drugs for medical uses.
  • the invention has particular utility in the metering and packaging of combinations of two or more pharmaceuticals and drugs for the same or co-morbid therapy, and will be described in connection with such utility / although other utilities are contemplated.
  • a pharmaceutical delivery package comprising fixed unit dose quantities of fixed quantities of two or more different active pharmaceutical ingredients (a) combined in a single delivery package,-and (b) segregated from one another within the package.
  • Previously direct placement of medication onto a substrate generally was Hmited to medical placement of large doses or required technology where the active pharmaceutical was mixed with the substrate or matrix to provide differential delivery, or coated with a material with desired release characteristics.
  • oiled-release is used to describe a system, i.e. method and materials for making an active ingredient available to the patient in accordance with a preselected condition, i.e. time, site, etc..
  • Controlled- release includes the use of instantaneous release, delayed release and sustained release.
  • Instantaneous release refers to immediate release to the patient.
  • Dellayed release means the active ingredient is not made available until some time delay after administration. Typically, dosages are administered by oral ingestion, although other forms of administration are contemplated in accordance with the present invention.
  • sustained release refers to release of active ingredient whereby the level of active ingredient available to the patient is maintained at some level over a period of time. The method of effecting each type of release can be varied. For example, the active-ingredient can be placed on a semi-permeable membrane having predetermined diffusion, dissolution, erosion or breakdown characteristics.
  • the active ingredient can be masked by a coating, a laminate, etc.
  • the present invention contemplates delivery of a controlled-release system which utilizes one or more of the "release” methods and materials.
  • the present invention advantageously can be employed in the development of multiple different release system(s).
  • SR sustained release
  • polymeric capsules For common methods of obtaining SR systems, see "Sustained and Controlled Release Drug Delivery Systems," Robinson, Joseph R., Ed., PP 138-171, 1978, Marcel Dekker, Inc. New York, NY.
  • SR sustained release
  • Heterogeneous matrices for example, compressed tablets, control the release of their therapeutic agents either by diffusion, erosion of the matrix or a combination of both.
  • Other SR systems focus on the fabrication of laminates of polymeric material and therapeutic agent which are then formed into a sandwich, relying on different diffusion or erosion rates to control release of the therapeutic agent.
  • Liquid-liquid encapsulation in a viscous syrup-like solution of polymer also has been known to be useful in controlling release of the therapeutic agent. Additionally, it is generally known that heterogeneous dispersions or solutions of therapeutic agents in water-swellable hydrogen matrices are useful in controlling the release of the agent by slow surface-to-center swelling of the matrix and subsequent diffusion of the agent from the water- swollen part of the matrix.
  • the dosage form During dissolution of a controlled-release matrix tablet, the dosage form generally remains as a non-disintegrating, slowly eroding entity from which the therapeutic agent leaches out, through a diffusion controlled process.
  • Conventional SR formulations are generally designed to release their active ingredients over an extended period of time, usually 8-24 hours.
  • Conventional SR formulations use waxes or hydrophilic gums as the primary drug carriers to prolong the release of the active ingredients.
  • Starch USP (potato or corn) is commonly used as a component in conventional tablet or hard shell capsule formulations.
  • the existing sustained release technologies generally involve relatively complicated formulations and manufacturing processes which often are difficult and expensive to precisely control.
  • OROS SR delivery system
  • OROS SR delivery system
  • the dosage unit should be able to deliver the system without interfering with its release .pattern.
  • controlled-release systems have been provided in the form of beads or particles which are packaged in a gelatin capsule for oral dosage. This method of delivery of the controlled-release system prevents damage to the coating on the beads.
  • Electrostatic charge has been employed to attract a given quantity of powder to a surface.
  • An example of this is the laser printer or the electrostatic copy device where a drum is charged and toner particles are attracted and held in position by the charge. The charge on the drum is neutralized by the attracted toner powder, thus limiting the amount of toner in accordance with the charge image on the drum. The charged powder on the printer drum is then transferred to a sheet of paper or other carrier to give a final image.
  • Patent 5,699,649 electrostatic charge technology is employed for transferring a predetermined amount of a finely powdered pharmaceutical or drug to a ' carrier or an intermediate such as a drum, carrying a charge of predetermined intensity and area, rotating the charged drum surface, carrying the predetermined amount of powdered pharmaceutical or drug on its surface, to a transfer station where the charge is overcome and the dry powder is transferred to a package which is then sealed.
  • a transfer station where the charge is overcome and the dry powder is transferred to a package which is then sealed.
  • a belt, or other movable surface is charged to a given potential in a localized area.
  • a predetermined amount of powdered pharmaceutical or drug may be deposited directly in a package using electrostatic charge technology.
  • the charge and area of charge can be determined experimentally for each dose of pharmaceutical or drug and each particle size distribution. This can be done by controlling either the charged area for a given charge density or the total electrostatic charge on any individual charged area. These conditions can be adjusted to provide essentially the exact desired amount of the particular pharmaceutical or drug to be transferred at the transfer station.
  • controlled quantities of powdered medication are formed in controlled release packages using electrostatic metering technology.
  • the present invention also provides, in another aspect, combination medication delivery systems in which the active ingredients are segregated from one another
  • Fig. 1 is a schematic flow diagram showing the various steps involved in practicing the present invention
  • Fig. 2 is an enlarged cross-sectional view of one embodiment of a controlled release tablet made in accordance with the present invention
  • Fig. 3 is a view, similar to Fig. 1, and showing alternative steps involved in practicing the present invention
  • Fig. 4 is a view, similar to Fig. 2, and showing an alternative form of a controlled release tablet made in accordance with the present invention
  • Fig.5 is a view similar to Fig.2, and showing yet another alternative embodiment of the present invention.
  • Fig. 6 is a view, similar to Fig. 2, and showing yet another embodiment of the invention.
  • Figs. 7 - 9 are views similar to Fig. 2, and showing yet other embodiments of the present invention.
  • Fig. 1 there is a schematic flow diagram of the various pieces of equipment needed to perform in the total process from powder supply to packaged pharmaceutical or drug, i.e. in controlled release tablet form, containing a specified amount of pharmaceutical or drug powder in the tablet or package.
  • the pharmaceutical or drug powder supply which is fed into a device 18 for creating an aerosol of the powder.
  • the powder particles are ionized at 20.
  • a carrier surface capable of maintaining a space charge on its surface. This can be a plastic belt, for example, or a selenium drum of the type used in Xerox TM photocopiers.
  • This carrier surface 24 is passed through a charging station 25 where a predetermined electrostatic charge 25 A (an electrostatic "image") is created on a predetermined area of the transfer surface.
  • This charged surface 25A then passes through a step 26 wherein powder is deposited on the carrier surface in a sufficient amount 26A to neutralize the charge carried by the carrier surface.
  • the carrier surface, carrying the predetermined amount 26A of powder on its surface is passed to a powder discharging device 30 which discharges the powder 26A from the surface 24 onto a membrane 29.
  • the powder may be placed directly onto the membrane 29.
  • the membrane 29 containing its charge of powder 26A then passes through a sealing step 32 wherein a second membrane 34 which may be porous, permeable or semi-permeable covers and seals the discharged powder 26A on the membrane 29. There is thus produced an aliquot of powdered medicine 26A sandwiched between semi-permeable or permeable membranes 29 and 34.
  • This sandwiched material is then passed to a cutting station 38 wherein the sandwich is cut into individual tablets or wafers 36.
  • the carrier surface with the electrostatic charge carries a known amount of charge on its surface and the polarity of this charge is opposite to that of the powder particles suspended in the chamber.
  • the charged particles migrate to the charged surface because of the attraction by the opposite nature of the charges. This migration of the particles continues until the charge on the carrier surface is neutralized.
  • the actual amount of powder mass transferred to the carrier surface is a function of the mass-to-charge ratio of the charged particles. Although it is difficult to achieve a linear relationship between the mass and the actual charge, it is possible to establish a fixed relationship between the surface area of the powder particles and the charge the powder particle is carrying at charge saturation. However, the surface area of a mixed group of powder particles of different sizes and shapes can be extremely difficult to calculate mathematically, particularly when the shapes are irregular, (e.g.
  • the simplest method of deterrnining the amount and area of charge to attract a given weight of particles is to estimate the correct area and charge and then apply the - estimated charge to the estimated area on the carrier surface 24 and expose this selectively charged area to a mass of powder which has been ionized in the ionizing step.
  • the amount of powder deposited can then be readily measured at the discharge step. Thereafter, either the size of the charged area or the amount of charge applied to the area at the charging station 25 can be adjusted upwardly or downwardly to provide the correct amount of charge, both in area and charge intensity, for picking up a desired weight of oppositely charged powder.
  • larger quantities of medication may be deposited.
  • a feature and advantage of the present invention is to produce carefully controlled doses of controlled release medication. Electrostatic metering and packaging as above described permits exact dosing. And, by employing selected porous, permeable or semi-permeable membranes for encapsulating the powdered medicine aliquots, drug release rate and also site of drug release can be determined by adjusting membrane material and/ or membrane thickness.
  • the membranes should be formed of ingestible materials having a selected permeability porosity to fluids at a selected site or sites within the alimentary canal, so as to permit controlled release of the medication.
  • one or both membranes 29., 34 may comprise acid-dissolvable materials when it is desired to release the medication into the stomach or the membranes 29, 34 may be alkaline-dissolvable materials at differing pH's to release into chosen locations within the intestine. Porosity, membrane thickness, etc., may be selected to provide desired rate of dissolution at the site of interest.
  • a two-component controlled release tablet 48 may be formed (see Fig.4) incorporating two different powdered medicines 50, 52, encapsulated between membranes 29 and 34 for simultaneous controlled release.
  • two different drugs 60, 62 may be layered on one another, separated by a membrane 64 so the two medications may be delivered sequentially either in the same location, or in different locations within the aUmentary canal.
  • Another feature and advantage of the multi-drug tablet of Fig. 4 and Fig. 5, as will be discussed in detail herein below, is that two normally incompatible drugs may be to be safely packaged in a single tablet.
  • the invention is susceptible to modification.
  • individual doses may be formed by electrostatic deposition in accordance with U.S. Patent No.5,714,007.
  • the tablet 70 may incorporate an adhesive layer 72 such as a mucosal adhesive, which in turn is covered by an acid or alkaline dissolvable protective membrane 74, which dissolves at a selected site allowing the adhesive to adhere, for example, to the intestinal wall, thereby increasing residence time of the medication in a chosen location.
  • an acid or alkaline activatable adhesive may be applied to the outer surface of the tablet.
  • the membrane may be a material which expands on contact with the acid or alkaline in the alimentary canal and becomes more porous whereby to slowly release medication in a chosen location within the alimentary canal.
  • a particular feature and advantage of the present invention is that it permits packaging, within a single tablet of two or more different drugs normally considered to be incompatible. Certain drugs are known to cause undesirable side effects which need to be countered by a second drug.
  • Omeprazole 1 which finds substantial utility as an oral antiulcer agent, also is known to block the release of B12 from its protein binding site in food. This can lead to pernicious anemia.
  • the present invention permits packaging of time-release Omeprazole with Vitamin B12 in an appropriate dosage of, e.g. 25 ⁇ gm - 1 mg. After taking the medication, one membrane will dissolve allowing absorption of the B12, while the remaining membrane package carrying the Omeprazole will pass into the small intestine where the drug is released and absorbed.
  • the invention is susceptible to modification.
  • the membranes have been described as being preformed, permeable, semiperrneable or porous material, one or both membranes could be formed in place from a gel or liquid.
  • the ability to accurately place the dose of medication onto a plurality of substrates and seal the dose with other membranes in accordance with the present invention allows for the fabrication of many different dosage forms; by altering the substrates and encapsulating material a single unit dose form can be fabricated with a plurality of different drugs in different coverings, membranes and barriers.
  • This will provide a single dosage form with multiple active ingredients each being delivered to the appropriate site for absorption.
  • two or more active medicaments may be combined in a single delivery container, i.e. pill, capsule or caplet without actually mixing the two or more ingredients.
  • the active ingredients are segregated from one another in a compartmentalized capsule 100.
  • two or more tablets 102, 104 each containing only one active ingredient could be placed in a larger absorbable capsule or encased in a larger tablet 106.
  • two or more active ingredients could each be formulated as encapsulated particles 108A, 108B, and the encapsulated mixed particles placed in a capsule 110 where the only contact is between the particle inert coatings, etc.
  • the combination of drugs of the present invention can be grouped into polypharmacy for a therapeutic area, and into polypharmacy for treatment of co-morbid diseases.
  • the invention will now be described with reference to the following non-lirniting examples.
  • Valacyclovir 2 and analogs and is used to treat Herpes Zoster. It is well known that two drugs Cimetidine 3 and Probenecid 4 both increase the AUC (area under curve) and Cmax. A combination drug can be constructed with a combination of either one or more of these components to provide more efficacy.
  • Enalapril 5 and analogs and isomers is an ACE inhibitor used for the treatment of hypertension.
  • This drug has been used with the following and analogs and isomers beta adrenegic-blocking agents, methyldopa, nitrate, calcium blocking agents, Hydralazine 6 , Prazosin 7 and Digoxin 8 without clinically significant side effects.
  • beta adrenegic-blocking agents beta adrenegic-blocking agents
  • methyldopa methyldopa
  • nitrate calcium blocking agents
  • Hydralazine 6 methyldopa
  • Prazosin 7 and Digoxin 8 without clinically significant side effects.
  • One or more of these agents may be combined with Enalapril to improve the compliance of patient with hypertension and hypertension and other cardiac diseases.
  • Ketoconazole 9 and analogs and isomers is used to treat fungal infections.
  • One of the side effects is the reduction of Testosterone. This side effect could be mitigated by the combination of Testosterone or one of its isomers or analogs to overcome the side effect.
  • Omeprazole 1 and analogs and isomers is also used in combination with Clarithoromycin 10 for ulcer treatment. These two drugs may be combined as a single dose for patient compliance.
  • Tamoxifen 11 and analogs and isomers used in treatment of breast cancer has a +/- 30% incident of water retention with weight gain > 5%. This can be a disturbing consequence for patients with an even more disturbing disease.
  • the addition of a diuretic or combination diuretic provides a single dosage form for reduction in side effect and compliance.
  • Isotretinoin 12 and analogs and isomers used for the treatment of postular acne has a severe danger if taken by a woman who is pregnant.
  • the incorporation of oral contraceptive medication eliminates the potential for pregnancy while medicated.
  • Metformin HO 13 and analogs and isomers are hypoglycemic agents which have been used in combination with Sulfonylurea 14 and analogs and isomers to treat Type 2 Diabetes. These two agents act in different ways on reducing glucose levels. A combination is helpful for those patients requiring more aggressive oral therapy for their diabetes.
  • Combinations for treating hypertension include: Combination # 1 Diuretic + Angiotensin converting enzyme inhibitor (ACE inhibitor)
  • An example includes the following classes of diuretics:
  • Carbonic anhydrase inhibitors - e.g. Dichlorophenamide 15 .
  • Loop diuretics e.g. Furosemide 16 .
  • a diuretic which is already formulated as a combination diuretic, e.g. Aldactazide, a combination of Spironolactone 20 (potassium sparing diuretic + hydrochlorothiazide).
  • This combination makes use of the different methods of action of two different diuretics separated by a barrier from an ACE inhibitor such as Enalapril maleate 21 , Fosinopril sodium 22 , or Lisinopril 23 .
  • Combination diuretics such as Zestoretic AstraZeneca a combination of Lisinopril 27 10 or 20 mg and Hydrochlorthiazide 17 12.5 or 25 mg, exist in tablet form comprising mixed active ingredients in the pill or tablet form.
  • the present invention segregates the Lisinopril and Hydrochlorthiazide.
  • Pills can be in the form of tablets, pills, capsules or other solid oral dosage forms.
  • Diuretics as described in combination drug # 1 plus an angiotensin II receptor antagonist such as Losartan potassium 24 and/ or Valsartan 25 .
  • a beta adrenergic blocking agent such as Bioprolol fumarate 26 or Metoprolol succinate 27 .
  • Combination # 5 Diuretic + Periferal Adrenergic Blocking Agent Diuretic as described in #11, plus a periferal adrenergic blocking agent such as: Prazosin hydrochloride 7 .
  • the drug barrier system of the present invention allows further drug combinations such as a Calcium chanel block combined with: beta blockers, ACE inhibitors, long acting nitrates, Digoxin 8 , oral hypoglycemic drugs as well as multiple combinations, and combinations with a diuretic and combination drugs # 2, 3 , 4 or more of the above-mentioned compounds.
  • the drug barrier system of the present invention also allows drug combinations such as ACE Inhibitors combined with Beta blockers, methyldopa nitrates, calcium channel blockers, Hydralazine 6 , Prazosin 7 , Digoxin 8 as well as multiple combinations, and combinations with a diuretic and combination drugs # 2, 3, 4 or more of the above-mentioned compounds.
  • drug combinations such as ACE Inhibitors combined with Beta blockers, methyldopa nitrates, calcium channel blockers, Hydralazine 6 , Prazosin 7 , Digoxin 8 as well as multiple combinations, and combinations with a diuretic and combination drugs # 2, 3, 4 or more of the above-mentioned compounds.
  • Biguanide such as Metaformin 13 with a sulfonylurea such as Glipizide 32 .
  • Biguanide such as Metaformin 13 with a thiazolidinedione such as Rosiglitazone maleate 33 .
  • HMG-CoA reductase inhibitor such as Simvastatin 35 , Atorvastatin 36 , or Pravastatin 37 with a bile acid sequestrant such as Colestipol hydrochloride 38 .
  • a HMG-CoA reductase inhibitor with a niacin compound A HMG-CoA reductase inhibitor with a niacin compound.
  • AntiMstamine plus Leukotriene modifier such as Loratadine 40 plus Montelukast 41 .
  • This example provides a drug combination for treating migraine.
  • This example provides a drug combination for treating postoperative/post-chemotherapy nausea.
  • Anti-nausea such as Doperidol 44 plus steroid such as Dexamethasone 45 .
  • This example provides various drug combinations for treating gastric/ duodenial ulcer.
  • SSRI Selective serotonin reuptake inhibitor
  • Protease inhibitor - ⁇ ndinayir (Crixivan 49 ) plus nuclear reverse transcriptase inhibitor - Efavirenz (Sustiva 50 ) " plus third drug, i.e.2 nd NRTT- Ziduvudine 51 or Azidothymidine 52 .
  • This example provides various drug combinations for treating anti-rejection cocktail after organ transplant.
  • Combination drug # 25 plus PPI/H2 for ulcer prevention - Omeprazole 1 Combination drug # 25 plus PPI/H2 for ulcer prevention - Omeprazole 1 .
  • This example provides a drug combination for treating infections with combination therapy such as tuberculosis.
  • combination drugs # 7-12 which are the combinations for control of the diabetes with any of combination drugs #1-7 or the single component medicaments used in the anti-hypertensive combinations.
  • a Calcium channel block such as Nifedipine 29 plus a vasodilator such as nitroglycerin.
  • This example provides a polypharmacy for treatment of seizure disorders.
  • a Gamma Aminobutyric analog such as Gabapentin 58 or a Gamma Aminobutyric stimulator such as Divalproex Sodium 59 plus a Benzodiazepine such as Alprazolam 60 .
  • An opioid and a non-opioid analgesic such as codeine and acetominophine.
  • Combination #34 An opioid and a bowel softener or evacuant.
  • This example provides polypharmacy for elimmating or minimizing gastric irritation caused by a primary drug.
  • a cyclooxygenase-2 inhibitor such as Celecoxib 61 plus Omeprazole 1 .
  • An anti-inflammatory such as Naproxen 62 plus Omeprazole 1 .
  • Prednisone 54 plus estrogen or progesterone to prevent bone mass loss. It is also possible to package two or more doses of the same active ingredient in slow and fast release forms.
  • This example provides polypharmacy for treating anxiety or panic disorder.
  • a selective serotonin reuptake inhibitor such as Paroxetine 63 plus a Benzodiazepine such as Lorazepam 64 .
  • An arninoketone such as Bupropion 65 plus Lorazepam 64 .
  • combination drugs including some of the above-listed combination drugs, also may be blended and packaged in a single tablet or capsule, when chemical interaction is not a problem.
  • the present invention also allows for the rapid production of different dosage medications using the same active ingredient, and allows for the development of medications with longer resident time.
  • Omeprazole 5-rnethoxy-2[[(4-methoxy-3,5-dimethyl-2-pyrindinly) methyl] sulfinyl]-lH-benzimidazole.
  • Valacyclovir L-valine, 2-[(2-amino-l,6-dihydro-6-oxo-9H-purin-9-yl) methoxy] ethyl ester, monohydrochlori.de.
  • Cimetidine N"-cyano-N-methyl-N , -[2-[[(5-methyl-l-H-imidazol-4-yl) methyl]thio]-ethyl]-quanidine.
  • Hydralazine 1-HydrazinophthaIazine monohydrochloride.
  • Prazosin HC1 hydrochoride salt of l-(4-amino-6,7-dimethoxy-2- quinazolinyl)-4- (2-f uroyl) piperazine .
  • Ketocanozole CIS - 1 - acetyl - 4[4 -[[2,4-dicheorophenyl -2-(lH- imidazol-1-ylmethyle) -l,3-dioxolan-4-yl] methocy] phenyl] piperazive.
  • Tamoxifen (Z)2-[4-(l,2-diphenyl-l-butenyl)phenoxy]-N, N- dimethylethanamine 2 hydroxy-l,2,3-propanetricarboxylate.
  • Isotretinoin 13-cis-retinoic acid.
  • Metformin N,N-dimemyHmidodcarbonimidic diamide hydrochloride.
  • Dichlorophenamide 4,5-dichloro-l,3- benzenedisulfonamide.
  • Furosemide 4-chloro-N-furfuryl-5-sulfamoylanthranilic acid.
  • Aldactone 17-hydroxy-7 ⁇ /]9/t -mercapto-3-oxo-17fl, 7 ⁇ g-pregn-4-ene-21- carboxylic acid gg g-lactone acetate.
  • Hydrochlorthiazide 6-chloro-3, 4-dihydro-2H-l, 2, 4-benzothiadiazine- 7-sulfonamide 1, 1-dioxide.
  • Chlorthalidone 2-Chloro-5-(2,3-dihydro-l-hydroxy-3-oxo-lH-isoindol- l-yl)benzenesulfonamide.
  • Spirolactone 1 -hy droxy-7flZp/za-mercapto-3-oxo-l 7al-ph -pregn-4-er ⁇ e- 21-carboxylic acid gamma -lactone acetate.
  • Fosinopril sodium L-proline, 4-cyclohexyl-l-[[[2-methyl-l-(l- oxopropoxy) propoxyl](4-phenylbutyl) phosphinyl]acetyl]-,sodium salt, trans-.
  • Lisinopril (S)-l-[N 2 -(l-Carboxy-3- phenylpropyl)-L-lysyl]-L-proline dihydrate.
  • Losartan potassium 2-butyl-4-chloro-l[p-(o-lH-tetrazol-5- ylphenyl)benzyl]imida ' zole-5-methanol monopotassium salt.
  • Valsartan as N- ⁇ - ⁇ xopenty ⁇ -N-tp'- ⁇ H-tetrazol-S-y ⁇ tlA'-biphenyl] ⁇ - yl]methyl]-L-valine.
  • Bioprolol fumarate ( ⁇ )-l-(4-((2-(l- Methylethoxy)emoxy)methyl) ⁇ henoxy)-3-((l-methyleti ⁇ yl)arnino)-2- propanol (E)-2-butenedioate (2:1) (salt).
  • Metoprolol succinate ( ⁇ ) l-(isopropylamino)-3-[p-(2-methoxyefhyl) phenoxy]-2-propanol succinate (2:1) (salt).
  • Amlodipine (R.S.) 3-ethyl-5-methyl-2-(2-arninoethoxymethyl)-4-(2- chloro ⁇ henyl)-l,4-dihyro-6-methyl-3,5-pyridinedicarboxylate benzenessulphonate.
  • Nifedipine 3,5-pyridinedicarboxylic acid, l,4-dihydro-2,6-dimethyl-4- (2-nitrophenyl)-,dimethyl ester.
  • Methyldopa levo-3-(3,4- dihydroxyphenyl)-2-methylalanine sesquihydrate.
  • Clonidine HCL (2,6-dic orophenylamino)-2- imidazoline hydrochloride.
  • Glucotrol l-cyclohexyl-3-[[p-(2-(5- methylpyrazinecarboxamido) ethyl]phenyl] sulf onyl]urea.
  • Rosiglitazone maleate (+)-5-[[4-[2-(methyl-2- pyridmylammo)ethoxy] ⁇ henyl]methyl]- 2,4-thiazolidinedione,(Z)-2- butenedioate.
  • Atorvastatin [R-(R*,R*)]-2-(4-fluorophenyl)-b,s-dihydroxy-5-(l- methylethyl)-3-phenyl-4 [(phenylamino)carbonyl]-lH-pyrrole-l- heptanoic acid, calcium salt (2:1) trihydrate.
  • Pravastatin l-Naphthalene-heptanoic acid, l,2,6,7,8,8a-hexahydro-b, d,6-trihydroxy-2-methyl -8-(2-methyl -1- oxobutoxy)-, monosodium salt,[lS-[la(bS*, d S*),2a,6a,8b(R*),8aa]]-.
  • Colestipol hydrochloride diethylenetriamine and 1 chloro-2,3- epoxypropane.
  • Loratadine ethyl4-(8-chloro-5,6-dihydro-llH-benzo[5,6]cyclohepta[l,2- &]pyridin-ll-yHdene)-l-piperidinecarboxylate.
  • Dexamethasone 9-fluoro-ll ⁇ ,17,21-trihydroxy- 16 ⁇ -methylpregna-l,4- diene-3,20-dione.
  • Famotidine l -(aminosulfonyl)-3-[[[2- [( ⁇ am omethylene)amino]-4- miazolyl]methyl]tMo]propanimidamide.
  • Buptopion ( ⁇ )-l-(3-chlorophenyl)-2-[(l,l-dimethylethyl)amino]-l- propanone hydrochloride.
  • Crixivan is [l(lS,2R),5(S)]-2,3,5-trideoxy-N-(2,3-dihydro-2- hydroxy- lH-mden-l-yl)-5-[2-[[(l,l-dimethylethyl)amino]carbonyl]-4-(3- ⁇ yridmy]n ethyl)- l-piperazinyl]-2-(phenylmethyl)-D-erythro- pentonamide sulfate (1:1) salt.
  • Azidothymidine 3'-azido-3'-deoxythymidine.
  • Cyclosporine [R-[RR*(E) ⁇ ] cyclic(L-alanyl-D-alanyl-N-methyl-L-leucyl- N-methyl-L-leucyl-N-methyl-L-valyl-3-hydroxy-N,4-dimethyl-L-2- amino-6-octenoyl-L- ⁇ -ammo-butyryl-N-methylglycyl-N-methyl-L- leucyl-L-valyl-N-methyl-L-leucyl).
  • Prednisone pregna-l,4-diene-3,ll,20-trione, 17,21-dihydroxy.
  • Isoniazid isonicotinic acid hydrazide.
  • Rifampin 3-(4-methyl-l-piperazinyl-iminom.ethyl)-rifamycin.
  • Gahapentin l-(aminomethyl)cyclohexanacetic acid.
  • Divalproex Sodium sodium hydrogen bis (2-pro ⁇ ylpentanoate).
  • Alprazolam 8-Chloro-l-methyl-6-phenyl-4H-s-triazolo [4,3-a][l,4] benzodiazepine.
  • Celecoxib 4-[5-(4-methylphenyl)-3- (trifluoromethyl)-lH-pyrazoH-yl] benzenesulfonamide.
  • Naproxen 2-naphthaleneacetic acid, 5 methoxy- a-methyl-,(+).
  • Bupropion ( ⁇ )-l-(3-chlorophenyl)-2-[(l,l-dirnethylethyl)amino]-l- propanone hydrochloride.

Abstract

Controlled quantities of powdered medication are formed in controlled release packages using electrostating metering. Also provided are combination medication therapy delivery packages comprising two or more active pharmaceuticals segregated from one another in a single delivery package.

Description

METERING AND PACKAGING OF CONTROLLED RELEASE MEDICATION
The present invention relates to the metering and packaging of precise quantities of pharmaceuticals and drugs for medical uses. The invention has particular utility in the metering and packaging of combinations of two or more pharmaceuticals and drugs for the same or co-morbid therapy, and will be described in connection with such utility/ although other utilities are contemplated.
The convenience of administering a single dose of a medication which releases multiple active ingredients in a controlled fashion and in a chosen location over an extended period of time, as opposed to the administration of a number of single doses at regular intervals, has long been recognized in the pharmaceutical arts. The advantage to the patient and clinician in having consistent and uniform blood levels of medication over an extended period .of time are likewise recognized. The advantages of a variety of controlled- release dosage forms are well known. Among the most important advantages are: (1) increased contact time for the drug to allow for local activity in the stomach, small intestine, colon, or other locus of activity; (2) increased and more efficient absorption for drugs which have specific absorption sites; (3) the ability to reduce the number of dosages per period of time; (4) employment of less total drug; (5) minimization or elimination of local and/ or systemic side effects; (6) minimization of drug accumulation associated with chronic dosing; (7) improved efficiency and safety of treatment; (8) reduced fluctuation of drug level; and (9) better patient compliance with overall disease management.
In accordance with the present invention there is provided a pharmaceutical delivery package comprising fixed unit dose quantities of fixed quantities of two or more different active pharmaceutical ingredients (a) combined in a single delivery package,-and (b) segregated from one another within the package. Additionally, many experts believe controlled release drug delivery has many important non-therapeutic ramifications as well, including a financial saving to the patient in terms of fewer lost work days, reduced hospitalization and fewer visits to the physician.
It is known that certain design parameters are critical to proper drug delivery. Typically, they are: (1) delivering the drug to the target tissue; (2) supplying the drug for a predetermined period of time; and (3) fabricating a delivery system that provides drug in the desired spatial and temporal pattern. Controlled release drug delivery systems are intended to utilize these parameters to achieve the aforementioned advantages as compared to conventional pharmaceutical dosing.
Previously direct placement of medication onto a substrate generally was Hmited to medical placement of large doses or required technology where the active pharmaceutical was mixed with the substrate or matrix to provide differential delivery, or coated with a material with desired release characteristics.
As used herein "contr oiled-release" is used to describe a system, i.e. method and materials for making an active ingredient available to the patient in accordance with a preselected condition, i.e. time, site, etc.. Controlled- release includes the use of instantaneous release, delayed release and sustained release. "Instantaneous release" refers to immediate release to the patient. "Delayed release" means the active ingredient is not made available until some time delay after administration. Typically, dosages are administered by oral ingestion, although other forms of administration are contemplated in accordance with the present invention. "Sustained release" refers to release of active ingredient whereby the level of active ingredient available to the patient is maintained at some level over a period of time. The method of effecting each type of release can be varied. For example, the active-ingredient can be placed on a semi-permeable membrane having predetermined diffusion, dissolution, erosion or breakdown characteristics.
Alternatively, the active ingredient can be masked by a coating, a laminate, etc. Regardless of the method of providing the desired release pattern, the present invention contemplates delivery of a controlled-release system which utilizes one or more of the "release" methods and materials. Moreover, the present invention advantageously can be employed in the development of multiple different release system(s).
The patent and scientific literature is replete with various sustained release (SR) methods and materials. For common methods of obtaining SR systems, see "Sustained and Controlled Release Drug Delivery Systems," Robinson, Joseph R., Ed., PP 138-171, 1978, Marcel Dekker, Inc. New York, NY. For example it is known to fill polymeric capsules with a solid, liquid, suspension or gel containing a therapeutic agent which is slowly released by diffusion through the capsule walls. Heterogeneous matrices, for example, compressed tablets, control the release of their therapeutic agents either by diffusion, erosion of the matrix or a combination of both. Other SR systems focus on the fabrication of laminates of polymeric material and therapeutic agent which are then formed into a sandwich, relying on different diffusion or erosion rates to control release of the therapeutic agent. Liquid-liquid encapsulation in a viscous syrup-like solution of polymer also has been known to be useful in controlling release of the therapeutic agent. Additionally, it is generally known that heterogeneous dispersions or solutions of therapeutic agents in water-swellable hydrogen matrices are useful in controlling the release of the agent by slow surface-to-center swelling of the matrix and subsequent diffusion of the agent from the water- swollen part of the matrix.
During dissolution of a controlled-release matrix tablet, the dosage form generally remains as a non-disintegrating, slowly eroding entity from which the therapeutic agent leaches out, through a diffusion controlled process. Conventional SR formulations are generally designed to release their active ingredients over an extended period of time, usually 8-24 hours. Conventional SR formulations use waxes or hydrophilic gums as the primary drug carriers to prolong the release of the active ingredients.
Starch USP (potato or corn) is commonly used as a component in conventional tablet or hard shell capsule formulations.
The existing sustained release technologies generally involve relatively complicated formulations and manufacturing processes which often are difficult and expensive to precisely control. For example, one well known SR delivery system, OROS, marketed by the Alza Corporation, involves laser drilling through a tablet to create passages for the release of the drug from the tablet core. In controlled release technologies, it is desirable to be able to incorporate the active ingredient in its controlled-release pattern in a single dosage unit without deteriorating the active ingredient. Moreover, the dosage unit should be able to deliver the system without interfering with its release .pattern.
Various methods have been devised to enable controlled-release systems to be delivered to a patient without destruction of the delivery system during manufacturing, handling and distribution. For example, controlled-release systems have been provided in the form of beads or particles which are packaged in a gelatin capsule for oral dosage. This method of delivery of the controlled-release system prevents damage to the coating on the beads.
Furthermore, when controlled-release active ingredients are incorporated in compression tablets, it may be difficult for many people to swallow such tablets. Moreover, dissolution of high compression tablets often initially is slow and erratic and may result in localized hot spots of alimentary tract irritation where disintegration and release of the active ingredient finally occurs. And, present systems do not allow for the accurate deposition of doses of powdered medication onto different substrates either in single packets, layered packet, or multipackets on the same plane of the base substrate. The present invention overcomes the disadvantages of the prior art by offering a simple and inexpensive means of incorporating active ingredient (the drug) with a multitude of controlled-release systems.
In our earlier U.S. Patent 5,699,649, granted December 23, 1997, we describe a method and apparatus for packaging microgram quantities of fine powders such as pharmaceuticals using electrostatic phototechnology techniques. More particularly, as described in our aforesaid U.S. Patent 5,699,649, the ability of powders to acquire an electrical charge advantageously is utilized for precisely measuring exact microgram quantities of the powder, whereupon these exact microgram quantities are then placed in individual containers, and the containers sealed.
Electrostatic charge has been employed to attract a given quantity of powder to a surface. An example of this is the laser printer or the electrostatic copy device where a drum is charged and toner particles are attracted and held in position by the charge. The charge on the drum is neutralized by the attracted toner powder, thus limiting the amount of toner in accordance with the charge image on the drum. The charged powder on the printer drum is then transferred to a sheet of paper or other carrier to give a final image. In our U.S. Patent 5,699,649, electrostatic charge technology is employed for transferring a predetermined amount of a finely powdered pharmaceutical or drug to a'carrier or an intermediate such as a drum, carrying a charge of predetermined intensity and area, rotating the charged drum surface, carrying the predetermined amount of powdered pharmaceutical or drug on its surface, to a transfer station where the charge is overcome and the dry powder is transferred to a package which is then sealed. In lieu of a drum, a belt, or other movable surface is charged to a given potential in a localized area. Alternatively, a predetermined amount of powdered pharmaceutical or drug may be deposited directly in a package using electrostatic charge technology.
When a given amount of a powdered pharmaceutical or drug is to be packaged, the charge and area of charge can be determined experimentally for each dose of pharmaceutical or drug and each particle size distribution. This can be done by controlling either the charged area for a given charge density or the total electrostatic charge on any individual charged area. These conditions can be adjusted to provide essentially the exact desired amount of the particular pharmaceutical or drug to be transferred at the transfer station.
In our U.S. Application Serial No. 09/097,104, we describe another electrostatic charge technology which may be adopted to be used for measuring and packaging unit doses of a pharmaceutical or drug in a readily ingestible form, i.e. as a tablet or capsule. The technology thus described also permits reproducible precise measurement and packaging of a pharmaceutical or drug, and which may be scaled from laboratory to pilot plant to full scale production without the need for recertification.
In accordance with one aspect of the present invention, controlled quantities of powdered medication are formed in controlled release packages using electrostatic metering technology. The present invention also provides, in another aspect, combination medication delivery systems in which the active ingredients are segregated from one another
Further features and objects of the present invention will become clear from the following detailed description taken in conjunction with the accompanying drawings, wherein like numerals depict like parts, and wherein:
Fig. 1 is a schematic flow diagram showing the various steps involved in practicing the present invention; Fig. 2 is an enlarged cross-sectional view of one embodiment of a controlled release tablet made in accordance with the present invention;
Fig. 3 is a view, similar to Fig. 1, and showing alternative steps involved in practicing the present invention;
Fig. 4 is a view, similar to Fig. 2, and showing an alternative form of a controlled release tablet made in accordance with the present invention;
Fig.5 is a view similar to Fig.2, and showing yet another alternative embodiment of the present invention;
Fig. 6 is a view, similar to Fig. 2, and showing yet another embodiment of the invention; and
Figs. 7 - 9 are views similar to Fig. 2, and showing yet other embodiments of the present invention.
Referring now to Fig. 1, there is a schematic flow diagram of the various pieces of equipment needed to perform in the total process from powder supply to packaged pharmaceutical or drug, i.e. in controlled release tablet form, containing a specified amount of pharmaceutical or drug powder in the tablet or package. At 16 is indicated the pharmaceutical or drug powder supply which is fed into a device 18 for creating an aerosol of the powder. Next the powder particles are ionized at 20. As will be indicated later, a number of these steps and pieces of equipment can be combined. At 24 is indicated a carrier surface capable of maintaining a space charge on its surface. This can be a plastic belt, for example, or a selenium drum of the type used in Xerox ™ photocopiers. This carrier surface 24 is passed through a charging station 25 where a predetermined electrostatic charge 25 A (an electrostatic "image") is created on a predetermined area of the transfer surface. This charged surface 25A then passes through a step 26 wherein powder is deposited on the carrier surface in a sufficient amount 26A to neutralize the charge carried by the carrier surface. Thereafter, the carrier surface, carrying the predetermined amount 26A of powder on its surface, is passed to a powder discharging device 30 which discharges the powder 26A from the surface 24 onto a membrane 29. Alternatively, the powder may be placed directly onto the membrane 29. The membrane 29 containing its charge of powder 26A, then passes through a sealing step 32 wherein a second membrane 34 which may be porous, permeable or semi-permeable covers and seals the discharged powder 26A on the membrane 29. There is thus produced an aliquot of powdered medicine 26A sandwiched between semi-permeable or permeable membranes 29 and 34.
This sandwiched material is then passed to a cutting station 38 wherein the sandwich is cut into individual tablets or wafers 36.
As mentioned previously in discussing Fig. 1, the carrier surface with the electrostatic charge carries a known amount of charge on its surface and the polarity of this charge is opposite to that of the powder particles suspended in the chamber. The charged particles migrate to the charged surface because of the attraction by the opposite nature of the charges. This migration of the particles continues until the charge on the carrier surface is neutralized.
The actual amount of powder mass transferred to the carrier surface is a function of the mass-to-charge ratio of the charged particles. Although it is difficult to achieve a linear relationship between the mass and the actual charge, it is possible to establish a fixed relationship between the surface area of the powder particles and the charge the powder particle is carrying at charge saturation. However, the surface area of a mixed group of powder particles of different sizes and shapes can be extremely difficult to calculate mathematically, particularly when the shapes are irregular, (e.g. non- spherical, microcrystalline, etc.) As mentioned ear Her, the simplest method of deterrnining the amount and area of charge to attract a given weight of particles is to estimate the correct area and charge and then apply the - estimated charge to the estimated area on the carrier surface 24 and expose this selectively charged area to a mass of powder which has been ionized in the ionizing step. The amount of powder deposited can then be readily measured at the discharge step. Thereafter, either the size of the charged area or the amount of charge applied to the area at the charging station 25 can be adjusted upwardly or downwardly to provide the correct amount of charge, both in area and charge intensity, for picking up a desired weight of oppositely charged powder. Likewise, using the technology of our co- pending application Serial No. 09/097,104, larger quantities of medication may be deposited.
A feature and advantage of the present invention is to produce carefully controlled doses of controlled release medication. Electrostatic metering and packaging as above described permits exact dosing. And, by employing selected porous, permeable or semi-permeable membranes for encapsulating the powdered medicine aliquots, drug release rate and also site of drug release can be determined by adjusting membrane material and/ or membrane thickness.
The membranes should be formed of ingestible materials having a selected permeability porosity to fluids at a selected site or sites within the alimentary canal, so as to permit controlled release of the medication. By way of example, one or both membranes 29., 34 may comprise acid-dissolvable materials when it is desired to release the medication into the stomach or the membranes 29, 34 may be alkaline-dissolvable materials at differing pH's to release into chosen locations within the intestine. Porosity, membrane thickness, etc., may be selected to provide desired rate of dissolution at the site of interest.
The inyention is susceptible to modification. For example, referring to Figs. 3 and 4 by adding a second powdered medicine supply and discharge station (shown generally at 40), a two-component controlled release tablet 48 may be formed (see Fig.4) incorporating two different powdered medicines 50, 52, encapsulated between membranes 29 and 34 for simultaneous controlled release.
Alternatively, as shown in Fig.5, two different drugs 60, 62 may be layered on one another, separated by a membrane 64 so the two medications may be delivered sequentially either in the same location, or in different locations within the aUmentary canal. Another feature and advantage of the multi-drug tablet of Fig. 4 and Fig. 5, as will be discussed in detail herein below, is that two normally incompatible drugs may be to be safely packaged in a single tablet.
The invention is susceptible to modification. For example, individual doses may be formed by electrostatic deposition in accordance with U.S. Patent No.5,714,007.
Other possibilities are possible. For example, referring to Fig. 6, the tablet 70 may incorporate an adhesive layer 72 such as a mucosal adhesive, which in turn is covered by an acid or alkaline dissolvable protective membrane 74, which dissolves at a selected site allowing the adhesive to adhere, for example, to the intestinal wall, thereby increasing residence time of the medication in a chosen location. Alternatively, an acid or alkaline activatable adhesive may be applied to the outer surface of the tablet. In yet another possibility, the membrane may be a material which expands on contact with the acid or alkaline in the alimentary canal and becomes more porous whereby to slowly release medication in a chosen location within the alimentary canal.
As mentioned above, a particular feature and advantage of the present invention is that it permits packaging, within a single tablet of two or more different drugs normally considered to be incompatible. Certain drugs are known to cause undesirable side effects which need to be countered by a second drug. For example, Omeprazole1 which finds substantial utility as an oral antiulcer agent, also is known to block the release of B12 from its protein binding site in food. This can lead to pernicious anemia. The present invention permits packaging of time-release Omeprazole with Vitamin B12 in an appropriate dosage of, e.g. 25μgm - 1 mg. After taking the medication, one membrane will dissolve allowing absorption of the B12, while the remaining membrane package carrying the Omeprazole will pass into the small intestine where the drug is released and absorbed.
The invention is susceptible to modification. For example, while the membranes have been described as being preformed, permeable, semiperrneable or porous material, one or both membranes could be formed in place from a gel or liquid.
The ability to accurately place the dose of medication onto a plurality of substrates and seal the dose with other membranes in accordance with the present invention, allows for the fabrication of many different dosage forms; by altering the substrates and encapsulating material a single unit dose form can be fabricated with a plurality of different drugs in different coverings, membranes and barriers. This will provide a single dosage form with multiple active ingredients each being delivered to the appropriate site for absorption. Alternatively, two or more active medicaments may be combined in a single delivery container, i.e. pill, capsule or caplet without actually mixing the two or more ingredients. For example, referring to Fig. 7, the active ingredients are segregated from one another in a compartmentalized capsule 100. Alternatively, two or more tablets 102, 104 each containing only one active ingredient, could be placed in a larger absorbable capsule or encased in a larger tablet 106. Or, as shown in Fig. 9, two or more active ingredients could each be formulated as encapsulated particles 108A, 108B, and the encapsulated mixed particles placed in a capsule 110 where the only contact is between the particle inert coatings, etc.
There are many drugs which could benefit from combinations to improve patient benefit. However, with many active ingredients, there is a question of chemical interaction. Thus, several drugs are normally prescribed as separate tablets or capsules which presents a problem in terms of patient compliance, e.g. TB triple therapy, AIDS multi-drug therapy, anti-infectives, etc. Also, delivery of two or more active medicaments could reduce side effects, and/ or improve therapeutic response which may in turn permit a decrease in the required dosage.
The combination of drugs of the present invention can be grouped into polypharmacy for a therapeutic area, and into polypharmacy for treatment of co-morbid diseases. The invention will now be described with reference to the following non-lirniting examples.
(1) Omeprazole1 and analogs and isomers - As noted above Omeprazole is an inhibitor of gastric secretion and also inhibits the absorption of certain drugs/ compounds that require stomach acid such as Vitamin B12, the deficit of which results in pernicious anemia. A combination of B12 with Omeprazole would eliminate the potential problem.
(2) Valacyclovir2 and analogs and is used to treat Herpes Zoster. It is well known that two drugs Cimetidine3 and Probenecid4 both increase the AUC (area under curve) and Cmax. A combination drug can be constructed with a combination of either one or more of these components to provide more efficacy.
(3) Enalapril5 and analogs and isomers is an ACE inhibitor used for the treatment of hypertension. This drug has been used with the following and analogs and isomers beta adrenegic-blocking agents, methyldopa, nitrate, calcium blocking agents, Hydralazine6, Prazosin7and Digoxin8 without clinically significant side effects. One or more of these agents may be combined with Enalapril to improve the compliance of patient with hypertension and hypertension and other cardiac diseases.
(4) Ketoconazole9 and analogs and isomers is used to treat fungal infections. One of the side effects is the reduction of Testosterone. This side effect could be mitigated by the combination of Testosterone or one of its isomers or analogs to overcome the side effect.
(5) Omeprazole1 and analogs and isomers is also used in combination with Clarithoromycin10 for ulcer treatment. These two drugs may be combined as a single dose for patient compliance.
(6) Tamoxifen11 and analogs and isomers used in treatment of breast cancer has a +/- 30% incident of water retention with weight gain > 5%. This can be a disturbing consequence for patients with an even more disturbing disease. The addition of a diuretic or combination diuretic provides a single dosage form for reduction in side effect and compliance.
(7) Isotretinoin12 and analogs and isomers used for the treatment of postular acne has a severe danger if taken by a woman who is pregnant. The incorporation of oral contraceptive medication eliminates the potential for pregnancy while medicated.
(8) Metformin HO13 and analogs and isomers are hypoglycemic agents which have been used in combination with Sulfonylurea14 and analogs and isomers to treat Type 2 Diabetes. These two agents act in different ways on reducing glucose levels. A combination is helpful for those patients requiring more aggressive oral therapy for their diabetes.
(9) This example provides various drug combinations for treating hypertension.
Combinations for treating hypertension include: Combination # 1 Diuretic + Angiotensin converting enzyme inhibitor (ACE inhibitor)
An example includes the following classes of diuretics:
1. Carbonic anhydrase inhibitors - e.g. Dichlorophenamide15.
2. Loop diuretics, e.g. Furosemide16.
3. Potassium sparing diuretics, e.g. Aldactone17. 4. Thiazides and related drugs, e.g. Hydrochlorthiazide18 and Chlorthalidone19.
5. A diuretic which is already formulated as a combination diuretic, e.g. Aldactazide, a combination of Spironolactone20 (potassium sparing diuretic + hydrochlorothiazide). This combination makes use of the different methods of action of two different diuretics separated by a barrier from an ACE inhibitor such as Enalapril maleate21, Fosinopril sodium22, or Lisinopril23.
Combination diuretics such as Zestoretic AstraZeneca a combination of Lisinopril2710 or 20 mg and Hydrochlorthiazide1712.5 or 25 mg, exist in tablet form comprising mixed active ingredients in the pill or tablet form. The present invention segregates the Lisinopril and Hydrochlorthiazide.
In accordance with the present invention, we can form e.g. 10 mg and 20 mg Lisinopril22 pills, and 12.5 and 25 mg Hydrochlorthiazide17 pills and then put them together with a barrier between two active ingredients. Pills can be in the form of tablets, pills, capsules or other solid oral dosage forms.
Combination # 2 Diuretic + Angiotensin II Receptor Antagonist
Diuretics as described in combination drug # 1 plus an angiotensin II receptor antagonist such as Losartan potassium24 and/ or Valsartan25.
These combinations also permit administration of two or more drugs which, if in direct contact, have an unacceptable reaction.
Combination # 3 Diuretic + Beta Adrenergic Blocking Agent
Diuretic as described in combination #1, plus a beta adrenergic blocking agent such as Bioprolol fumarate26 or Metoprolol succinate27.
Combination # 4 Diuretic + Calcium chanel block
Diuretic as described in combination #1, plus a Calcium chanel block such as Amlodipine28 or Nifedipine29.
Combination # 5 Diuretic + Periferal Adrenergic Blocking Agent Diuretic as described in #11, plus a periferal adrenergic blocking agent such as: Prazosin hydrochloride7.
Combination # 6 Diuretic + Adrenergic central stimulant
Diuretic as described in #1, plus an adrenergic central stimulant such as: Methyldopa30 or Clonidine31.
Combination # 7 Diuretic + Endothelin A
This is a new class of drugs.
The drug barrier system of the present invention allows further drug combinations such as a Calcium chanel block combined with: beta blockers, ACE inhibitors, long acting nitrates, Digoxin8, oral hypoglycemic drugs as well as multiple combinations, and combinations with a diuretic and combination drugs # 2, 3 , 4 or more of the above-mentioned compounds.
Combination #8 ACE Inhibitors + Beta Blockers
The drug barrier system of the present invention also allows drug combinations such as ACE Inhibitors combined with Beta blockers, methyldopa nitrates, calcium channel blockers, Hydralazine6, Prazosin7, Digoxin8 as well as multiple combinations, and combinations with a diuretic and combination drugs # 2, 3, 4 or more of the above-mentioned compounds.
(10) This example provides various drug combinations for treating diabetes.
Combination # 9
Biguanide such as Metaformin13 with a sulfonylurea such as Glipizide32.
Combination # 10
Biguanide such as Metaformin13 with a thiazolidinedione such as Rosiglitazone maleate33.
Combination # 11
Metaformin13 with an alpha glucosidase inhibitor such as Cerivastatin34. Combination # 12
Short acting oral insulin with sustained release oral insulin.
(11) This example provides various drug combinations for treating hyperlipidemia.
Combination # 13
HMG-CoA reductase inhibitor such as Simvastatin35, Atorvastatin36, or Pravastatin37 with a bile acid sequestrant such as Colestipol hydrochloride38.
Combination # 14
A HMG-CoA reductase inhibitor with a niacin compound.
Combination # 15
A HMG-CoA reductase inhibitor or combination #14 with a hypolipidemia agent such as Gemfibrozil39.
(12) This example provides various drug combinations for treating congestive heart failure.
Combination # 16
Digitalis plus an ACE inhibitor with or without a diuretic, and optionally including a beta blocker. Combination # 17 Digitalis plus any of the combination drugs #l-#7.
(13) This example provides various drug combinations for treating asthma/ allergy.
Combination # 18
Rapid onset anti-histamine plus sustained release anti-histamine. Combination # 19
AntiMstamine plus Leukotriene modifier, such as Loratadine40 plus Montelukast41.
(14) This example provides a drug combination for treating migraine.
Combination # 20 Rapid acting 5-HT1 receptor agonist such as Naratriptin HC142 plus a long acting 5-HT1 receptor agonist such as Sumatriptan43.
(15) This example provides a drug combination for treating postoperative/post-chemotherapy nausea.
Combination # 21
Anti-nausea such as Doperidol44 plus steroid such as Dexamethasone45.
(16) This example provides various drug combinations for treating gastric/ duodenial ulcer.
Combination # 22
Quick onset H blocker such as Farnotidine46 plus a proton pump inhibitor such as Omeprazole1.
Combination # 3
Selective serotonin reuptake inhibitor (SSRI) fluoxetine (Prozac47) and Aminoketon - Buptopion48.
(17) This example provides a drug combination for treating HIV. Combination # 24
Protease inhibitor - ϊndinayir (Crixivan49) plus nuclear reverse transcriptase inhibitor - Efavirenz (Sustiva50)"plus third drug, i.e.2nd NRTT- Ziduvudine51 or Azidothymidine52.
(18) This example provides various drug combinations for treating anti-rejection cocktail after organ transplant.
Combination # 25
Cyclosporine53 plus steroid - Prednisone54. Combination # 26
Combination drug # 25 plus PPI/H2 for ulcer prevention - Omeprazole1.
(19) This example provides a drug combination for treating infections with combination therapy such as tuberculosis.
Combination #27 Triple combination Isoniazid55 and Pyrazidamide56 and Rifampin57.
(20) This example provides a polypharmacy for treatment of co- morbid diseases.
Combination # 28
80% + of diabetics are also hypertensive. Therefore a combination of any of combination drugs # 7-12 which are the combinations for control of the diabetes with any of combination drugs #1-7 or the single component medicaments used in the anti-hypertensive combinations.
Combination # 29
Hyperlipidemia is frequently concurrent with cardiac disease therefore any of combination drugs # 13-17 plus any of combination drugs # 1-7.
(21) This example provides a polypharmacy for treatment of Angina. Combination #30
A Calcium channel block such as Nifedipine29 plus a vasodilator such as nitroglycerin.
(22) This example provides a polypharmacy for treatment of seizure disorders.
Combination #31 . A Gamma Aminobutyric analog such as Gabapentin58 or a Gamma Aminobutyric stimulator such as Divalproex Sodium59 plus a Benzodiazepine such as Alprazolam60.
(23) This example provides various drug combinations for treating pain and the side effects of opioids:
Combination #32
An opioid and a non-opioid analgesic such as codeine and acetominophine.
Combination #33
An opioid and an antiemetic.
Combination #34 An opioid and a bowel softener or evacuant.
(24) This example provides polypharmacy for elimmating or minimizing gastric irritation caused by a primary drug.
Combination #35
A cyclooxygenase-2 inhibitor such as Celecoxib61 plus Omeprazole1.
Combination #36
An anti-inflammatory such as Naproxen62 plus Omeprazole1.
(25) This example provides polypharmacy for countering the effect of long term use of Prednisone54.
Combination #37
Prednisone54 plus testosterone to prevent muscle mass loss. Combination #38
Prednisone54 plus estrogen or progesterone to prevent bone mass loss. It is also possible to package two or more doses of the same active ingredient in slow and fast release forms.
(26) This example provides polypharmacy for treating anxiety or panic disorder.
Combination #39
A selective serotonin reuptake inhibitor such as Paroxetine63 plus a Benzodiazepine such as Lorazepam64.
Combination #40
An arninoketone such as Bupropion65 plus Lorazepam64.
Various analogs and isomers of the foregoing drugs also advantageiously may be employed.
It should be noted that certain combination drugs, including some of the above-listed combination drugs, also may be blended and packaged in a single tablet or capsule, when chemical interaction is not a problem. The present invention also allows for the rapid production of different dosage medications using the same active ingredient, and allows for the development of medications with longer resident time.
APPENDIX
1. Omeprazole: 5-rnethoxy-2[[(4-methoxy-3,5-dimethyl-2-pyrindinly) methyl] sulfinyl]-lH-benzimidazole.
2. Valacyclovir: L-valine, 2-[(2-amino-l,6-dihydro-6-oxo-9H-purin-9-yl) methoxy] ethyl ester, monohydrochlori.de.
3. Cimetidine: N"-cyano-N-methyl-N,-[2-[[(5-methyl-l-H-imidazol-4-yl) methyl]thio]-ethyl]-quanidine.
4. Probenecid: 4-[(dipropylamino) sulfonyl] benzoic acid (molecular weigh 285.36).
5. Enalapril: (S)-l-[N-[l-(ethoxycarbonyl)-3-phenylpropyl]-L-alanyl]-L- proline, (Z)-2-butenedioate salt.
6. Hydralazine: 1-HydrazinophthaIazine monohydrochloride.
7. Prazosin HC1: hydrochoride salt of l-(4-amino-6,7-dimethoxy-2- quinazolinyl)-4- (2-f uroyl) piperazine .
8. Digoxin:. 3B-[(o-2,6-dideoxy-B-D-ribo-hexopyranosyl-(iri4)-0-2, 6- dideoxy-B-D-ribo-hexopyranosyl - (Hl4)-2,6-dideoxy -B-D-ribo- hexopyranosyl) oxy] -12B, 14-dihydroxy-5B-card-20(22) enolide.
9. Ketocanozole: CIS - 1 - acetyl - 4[4 -[[2,4-dicheorophenyl -2-(lH- imidazol-1-ylmethyle) -l,3-dioxolan-4-yl] methocy] phenyl] piperazive.
10. Clarithoromycin: 6-0-methylerythromycin.
11. Tamoxifen: (Z)2-[4-(l,2-diphenyl-l-butenyl)phenoxy]-N, N- dimethylethanamine 2 hydroxy-l,2,3-propanetricarboxylate.
12. Isotretinoin: 13-cis-retinoic acid.
13. Metformin: N,N-dimemyHmidodcarbonimidic diamide hydrochloride.
14. Sulfonylurea: l-[[P-[2-(5-chloro-o-anisamido) ethyl] phenyl] sulfonyl-]- 3-cyclohexylure. .
15. Dichlorophenamide: 4,5-dichloro-l,3- benzenedisulfonamide. 16. Furosemide: 4-chloro-N-furfuryl-5-sulfamoylanthranilic acid.
17. Aldactone: 17-hydroxy-7α/]9/t -mercapto-3-oxo-17fl, 7ι g-pregn-4-ene-21- carboxylic acid gg g-lactone acetate.
18. Hydrochlorthiazide: 6-chloro-3, 4-dihydro-2H-l, 2, 4-benzothiadiazine- 7-sulfonamide 1, 1-dioxide.
19. Chlorthalidone: 2-Chloro-5-(2,3-dihydro-l-hydroxy-3-oxo-lH-isoindol- l-yl)benzenesulfonamide.
20. Spirolactone: 1 -hy droxy-7flZp/za-mercapto-3-oxo-l 7al-ph -pregn-4-erιe- 21-carboxylic acid gamma -lactone acetate.
21. Enalapril maleate: (S)-l-[N-[l-(ethoxycarbonyl) -3-phenylpropyl]-L- alanyl] -L-proline, (Z)-2-butenedioate salt (1:1).
22. Fosinopril sodium: L-proline, 4-cyclohexyl-l-[[[2-methyl-l-(l- oxopropoxy) propoxyl](4-phenylbutyl) phosphinyl]acetyl]-,sodium salt, trans-.
23: Lisinopril: (S)-l-[N2-(l-Carboxy-3- phenylpropyl)-L-lysyl]-L-proline dihydrate.
24. Losartan potassium: 2-butyl-4-chloro-l[p-(o-lH-tetrazol-5- ylphenyl)benzyl]imida'zole-5-methanol monopotassium salt.
25. Valsartan: as N-^-όxopenty^-N-tp'-^H-tetrazol-S-y^tlA'-biphenyl]^- yl]methyl]-L-valine.
26. Bioprolol fumarate: (±)-l-(4-((2-(l- Methylethoxy)emoxy)methyl)ρhenoxy)-3-((l-methyletiιyl)arnino)-2- propanol (E)-2-butenedioate (2:1) (salt).
27. Metoprolol succinate: (±) l-(isopropylamino)-3-[p-(2-methoxyefhyl) phenoxy]-2-propanol succinate (2:1) (salt).
28. Amlodipine: (R.S.) 3-ethyl-5-methyl-2-(2-arninoethoxymethyl)-4-(2- chloroρhenyl)-l,4-dihyro-6-methyl-3,5-pyridinedicarboxylate benzenessulphonate.
29. Nifedipine: 3,5-pyridinedicarboxylic acid, l,4-dihydro-2,6-dimethyl-4- (2-nitrophenyl)-,dimethyl ester. 30. Methyldopa: levo-3-(3,4- dihydroxyphenyl)-2-methylalanine sesquihydrate.
31. Clonidine HCL: (2,6-dic orophenylamino)-2- imidazoline hydrochloride.
32. ' Glucotrol: l-cyclohexyl-3-[[p-(2-(5- methylpyrazinecarboxamido) ethyl]phenyl] sulf onyl]urea.
33. Rosiglitazone maleate: (+)-5-[[4-[2-(methyl-2- pyridmylammo)ethoxy]ρhenyl]methyl]- 2,4-thiazolidinedione,(Z)-2- butenedioate.
34. Cerivastatin: [S-[ R* S'-( E)] -7-[ 4-( 4-b fluorophenyl)-5- methoxymethyl)- 2,6bis( 1-met ylethyl) 3-pyridinyll-3,5-dihydroxy+ heptenoate.
35. Simvastatin^^-dimethyHl^^/^δ^a-hexahydro-S^-dimethyl-δ-p- (tetrahydro-4-hydroxy-6-oxo-2H-ρyran-2-yl)-ethyl]-l-naphthalenyl ester, [lS*-[la,3a,7b,8b(2S*,4S),-8ab]] .
36. Atorvastatin: [R-(R*,R*)]-2-(4-fluorophenyl)-b,s-dihydroxy-5-(l- methylethyl)-3-phenyl-4 [(phenylamino)carbonyl]-lH-pyrrole-l- heptanoic acid, calcium salt (2:1) trihydrate.
37. Pravastatin: l-Naphthalene-heptanoic acid, l,2,6,7,8,8a-hexahydro-b, d,6-trihydroxy-2-methyl -8-(2-methyl -1- oxobutoxy)-, monosodium salt,[lS-[la(bS*, d S*),2a,6a,8b(R*),8aa]]-.
38. Colestipol hydrochloride: diethylenetriamine and 1 chloro-2,3- epoxypropane.
39. Gemfibrozil: 5- (2,5-dimethylphenoxy)-2,2-dimethylpentanoic acid.
40. Loratadine: ethyl4-(8-chloro-5,6-dihydro-llH-benzo[5,6]cyclohepta[l,2- &]pyridin-ll-yHdene)-l-piperidinecarboxylate.
41. Montelukast: [R-(E)]-l-[[[l-[3-[2-(7-chloro-2- quinolinyl)ethenyl]phenyl]-3-[2-(l-hydroxy-l- methylethyl)phenyl]propyl]thio]methyl]cyclopropaneacetic acid, monosodium salt.
42. Naratriptin HCL: N-methyl-3-( l-methyl-4-piperidinyl)-lH-indole-5- ethanesulfonamide monohydrochloride. 43. Sumatriptan: 3-[2-(mmethylan ino)ethyl]-N-methyl-indole -5- methanesulfonamide succinate (1:1).
44. Doperidol: l-(l-[3-(p-fluorobenzoyl)propyl]-l,2,3,6-tetrahydro-4- pyridyl)-2-benziimdazolrnone.
45. Dexamethasone: 9-fluoro-ll β,17,21-trihydroxy- 16α-methylpregna-l,4- diene-3,20-dione.
46. Famotidine: l-(aminosulfonyl)-3-[[[2- [(ώam omethylene)amino]-4- miazolyl]methyl]tMo]propanimidamide.
47. Prozac: (±)-N-methyl-3-phenyl-3-[(α,α,α-trifluoro-p-tolyl)- oxy]propylamine hydrochloride.
48. Buptopion: (±)-l-(3-chlorophenyl)-2-[(l,l-dimethylethyl)amino]-l- propanone hydrochloride.
49. Crixivan: is [l(lS,2R),5(S)]-2,3,5-trideoxy-N-(2,3-dihydro-2- hydroxy- lH-mden-l-yl)-5-[2-[[(l,l-dimethylethyl)amino]carbonyl]-4-(3- ρyridmy]n ethyl)- l-piperazinyl]-2-(phenylmethyl)-D-erythro- pentonamide sulfate (1:1) salt.
50. Sustiva: (S)-6-chloro-4-(cyclopropylethynyl)-l,4-dihydro-4- (trifluoromethyl)-2H-3,l-benzoxazin-2-one.
51. Ziduvudine: 3'-azido-3'-deoxythymidine.
52. Azidothymidine: 3'-azido-3'-deoxythymidine.
53. Cyclosporine: [R-[RR*(E)}] cyclic(L-alanyl-D-alanyl-N-methyl-L-leucyl- N-methyl-L-leucyl-N-methyl-L-valyl-3-hydroxy-N,4-dimethyl-L-2- amino-6-octenoyl-L-α-ammo-butyryl-N-methylglycyl-N-methyl-L- leucyl-L-valyl-N-methyl-L-leucyl).
54. Prednisone: pregna-l,4-diene-3,ll,20-trione, 17,21-dihydroxy.
55. Isoniazid: isonicotinic acid hydrazide.
56. Pyrazinamide: pyrazinecarboxamide.
57. Rifampin: 3-(4-methyl-l-piperazinyl-iminom.ethyl)-rifamycin. 58. Gahapentin: l-(aminomethyl)cyclohexanacetic acid.
59. Divalproex Sodium: sodium hydrogen bis (2-proρylpentanoate).
60. Alprazolam: 8-Chloro-l-methyl-6-phenyl-4H-s-triazolo [4,3-a][l,4] benzodiazepine.
61. Celecoxib: 4-[5-(4-methylphenyl)-3- (trifluoromethyl)-lH-pyrazoH-yl] benzenesulfonamide.
62. Naproxen: 2-naphthaleneacetic acid, 5 methoxy- a-methyl-,(+).
63. Paroxetine aOailable as Immediate-Release Tablets and Oral Suspension as: (-)-frflMS-4R-(4,-fluorophenyl)-3S-[(3l,4'-methylenedioxyρhenoxy) methyl] piperidine hydrochloride hemihydrate and as Controlled- Release Tablets as: (-) - (3S,4R)-4-[(ρ-fluorophenyl)-3-[(3,4- methylenedioxy) phenoxy]methyl]piperidine hydrochloride hemihydrate.
64. Lorazepam: 7-chloro-5-(0-chlorophenyl)-l,3-dihydro-3-hydroxy-2H- l,4-benzo-diazepin-2-one.
65. Bupropion: (±)-l-(3-chlorophenyl)-2-[(l,l-dirnethylethyl)amino]-l- propanone hydrochloride.

Claims

1. A controlled release pharmaceutical delivery package comprising a unit aliquot dose of a pharmaceutical electrostatically deposited on a porous, permeable or semi-permeable ingestible membrane.
2. A pharmaceutical delivery package according to claim 1, wherein said membrane comprises an acid-dissolvable material.
3. A pharmaceutical delivery package according to claim 1, wherein said membrane comprises an alkali-dissolvable material.
4. A pharmaceutical delivery package according to claim 1, and comprising two or more pharmaceuticals deposited on said membrane, and separated by one another by one or more barriers or membranes.
5. A pharmaceutical delivery package according to claim 1, and further comprising an adhesive on the outer surface of the membrane.
6. A pharmaceutical delivery package according to claim 5, wherein the adhesive is aςid or alkylene activatable.
7. A pharmaceutical delivery package according to claim 5, and further comprising an alkali or acidic dissolvable membrane covering the adhesive.
8. A pharmaceutical delivery package according to claim 1, wherein said membrane comprises a material which expands upon contact with acid or alkaline in the alimentary canal, whereby to become more porous.
9. A pharmaceutical delivery package comprising two or more active pharmaceuticals (a) combined in a single delivery package, and (b) segregated from one another, wherein said single delivery package comprises an unitary structure for repeatable administration of fixed quantifies of said two or more active ingredients to the user.
10. A pharmaceutical delivery package according to claim 9, wherein said active ingredients are segregated from one another in a compartmentalized capsule.
11. A pharmaceutical delivery package according to claim 9, wherein said pharmaceuticals are segregated from one another in a tablet.
12. A pharmaceutical delivery package according to claim 9, wherein said pharmaceuticals are encapsulated within inert coatings.
13. A pharmaceutical delivery package comprising a combination of Ketoconazole and testosterone.
14. A pharmaceutical delivery package comprising a combination of Valacylovir and one or both of Cimetidine and Probenecid.
15. A pharmaceutical delivery package comprising a combination of Enalapril and a beta adrenergic-blocking agent, methyldopa, nitrate, a calcium blocking agent, hydrazine, Prazosin or Digoxin.
16. A pharmaceutical delivery package comprising a combination of Omeprazole and B12.
17. A pharmaceutical delivery package comprising a combination of Omeprazole and Clarithoromycin.
18. A pharmaceutical delivery package comprising a combination of Tamoxifen and a diuretic.
19. A pharmaceutical delivery package comprising a combination of Isotretinoin and an oral contraceptive.
20. A pharmaceutical delivery package comprising a combination of Metformin HCI and Solfonylurea.
21. A pharmaceutical delivery package comprising a combination of a diuretic and an Angiotensin converting enzyme inhibitor (ACE inhibitor).
22. A pharmaceutical delivery package comprising a combination of a diuretic and an Angiotensin II Receptor Antagonist.
23. A pharmaceutical delivery package comprising a combination of a diuretic and a Beta Adrenergic Blocking Agent.
24. A pharmaceutical delivery package comprising a combination of a diuretic and a Calcium channel block.
25. . A pharmaceutical delivery package comprising a combination of a diuretic and a Periferal Adrenergic Blocking Agent.
26. A pharmaceutical delivery package comprising a combination of a diuretic and an Adrenergic central stimulant.
27. A pharmaceutical delivery package comprising a combination of a diuretic and Endothelin A.
28. A pharmaceutical delivery package comprising a combination of an ACE inhibitor and a beta blocker.
29. A pharmaceutical delivery package comprising a combination of a biguanide and a sulfonylurea.
30. A pharmaceutical delivery package comprising a combination of a biguanide and a thiazolidinedione.
31. A pharmaceutical delivery package comprising a combination of Metaformin and an alpha glucosidase inhibitor.
32. A pharmaceutical delivery package comprising a combination of a short acting oral insulin with a sustained release oral insulin.
33. A pharmaceutical delivery package comprising a combination of an HMG-CoA reductase inhibitor with a bile acid sequestrant.
34. A pharmaceutical delivery package comprising a combination of an HMG-CoA reductase inhibitor with a niacin compound.
35. A pharmaceutical delivery package comprising a combination of an HMG-CoA reductase inhibitor with a hypolipidemia agent.
36. A pharmaceutical delivery package comprising a combination of an HMG-CoA reductase inhibitor, a niacin compound and a hypolipidemia agent.
37. A pharmaceutical delivery package comprising a combination of digitalis plus an ACE inhibitor.
38. A pharmaceutical delivery package comprising a combination of digitalis plus an ACE inhibitor and a diuretic.
39. A pharmaceutical delivery package comprising a combination of digitalis plus an ACE inhibitor, a diuretic and a beta blocker.
40. A pharmaceutical delivery package comprising a combination of a rapid onset anti-Mstamine plus a sustained release anti-histamine.
41. A pharmaceutical delivery package comprising a combination of an antiWstamine plus a Leukotriene modifier.
42. A pharmaceutical delivery package comprising a combination of a rapid acting 5-HT1 receptor agonist plus a long acting 5-HT1 receptor agonist.
43. A pharmaceutical delivery package comprising a combination of an anti-nausea plus a steroid.
44. A pharmaceutical delivery package comprising a combination of - a quick onset H blocker plus a proton pump inhibitor.
45. A pharmaceutical delivery package comprising a combination of a selective serotonin reuptake inhibitor (SSRI) fluoxetine and an Aminoketon.
46. A pharmaceutical delivery package comprising a combination of a protease inhibitor plus a nuclear reverse transcriptase inhibitor plus 2nd NRTI-Ziduvudine or Azidothymidine.
47. A pharmaceutical delivery package comprising a combination of cyclosporine plus a steroid.
48. A pharmaceutical delivery package comprising a combination of cyclosporine plus a steroid, plus a PPI/H2.
49. A pharmaceutical delivery package comprising a combination of Isoniazid, Pyrazidamide and Rifampin.
50. A pharmaceutical delivery package comprising a combination of a Calcium channel block plus a vasodilator.
51. A pharmaceutical delivery package comprising a combination of a Gamma Aminobutyric analog or a Gamma Aminobutyric stimulator plus a Benzodiazepine.
52. A pharmaceutical delivery package comprising a combination of an opioid and a non-opioid analgesic.
53. A pharmaceutical delivery package comprising a combination of an opioid and an antiemetic.
54. A pharmaceutical delivery package comprising a combination of an opioid and a bowel softener or evacuant.
55. A pharmaceutical delivery package comprising a combination of a cyclooxygenase-2 inhibitor plus Omeprazole.
56. A pharmaceutical delivery package comprising a combination of an anti-inflammatory plus' Omeprazole.
57. A pharmaceutical delivery package comprising a combination of -prednisone plus testosterone.
58. A pharmaceutical delivery package comprising a combination of prednisone plus estrogen.
59. A pharmaceutical delivery package comprising a combination of a selective serotonin reuptake inhibitor plus a benzodiazepine.
60. A pharmaceutical delivery package comprising a combination of an aminoketone plus Lorazepam.
61. A pharmaceutical delivery package as claimed in any one of claims 13 to 19, and further including digitalis.
62. A pharmaceutical delivery package as claimed in any one of claims 20 to 24, and further including a combination of any of the combinations of any one of claims 13 to 19.
63. A pharmaceutical delivery package as claimed in any one of claims 25 to 29, and further including a combination of any of the combinations of any one of claims 13 tol9.
PCT/US2002/016185 2001-05-31 2002-05-22 Metering and packaging of controlled release medication WO2002096347A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US10/479,438 US20040156903A1 (en) 2002-05-22 2002-05-22 Metering and packaging of controlled release medication
CA002448997A CA2448997A1 (en) 2001-05-31 2002-05-22 Metering and packaging of controlled release medication
EP02731902A EP1408907A4 (en) 2001-05-31 2002-05-22 Metering and packaging of controlled release medication
NZ529696A NZ529696A (en) 2001-05-31 2002-05-22 Metering and packaging of controlled release medication
BRPI0209720A BRPI0209720A2 (en) 2001-05-31 2002-05-22 dosage and packaging of controlled release medication
JP2002592860A JP2005514966A (en) 2001-05-31 2002-05-22 Controlled release drug metering and packaging
AU2002303840A AU2002303840A1 (en) 2001-05-31 2002-05-22 Metering and packaging of controlled release medication
US11/549,492 US20070087048A1 (en) 2001-05-31 2006-10-13 Oral dosage combination pharmaceutical packaging

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29478601P 2001-05-31 2001-05-31
US60/294,786 2001-05-31

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/549,492 Continuation-In-Part US20070087048A1 (en) 2001-05-31 2006-10-13 Oral dosage combination pharmaceutical packaging

Publications (2)

Publication Number Publication Date
WO2002096347A2 true WO2002096347A2 (en) 2002-12-05
WO2002096347A3 WO2002096347A3 (en) 2003-03-27

Family

ID=23134944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/016185 WO2002096347A2 (en) 2001-05-31 2002-05-22 Metering and packaging of controlled release medication

Country Status (8)

Country Link
EP (1) EP1408907A4 (en)
JP (1) JP2005514966A (en)
AU (1) AU2002303840A1 (en)
BR (1) BRPI0209720A2 (en)
CA (3) CA2614902A1 (en)
NZ (1) NZ529696A (en)
WO (1) WO2002096347A2 (en)
ZA (1) ZA200402466B (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004010978A1 (en) 2002-07-25 2004-02-05 Glaxo Group Limited Multicomponent pharmaceutical dosage form
WO2007047371A2 (en) * 2005-10-14 2007-04-26 Microdose Technologies, Inc. Pharmaceutical packaging of an oral dosage combination
US7404968B2 (en) 1999-08-18 2008-07-29 Microdose Technologies, Inc. Metering and packaging of controlled release medication
US7670612B2 (en) 2002-04-10 2010-03-02 Innercap Technologies, Inc. Multi-phase, multi-compartment capsular delivery apparatus and methods for using same
US20100260669A1 (en) * 2004-05-13 2010-10-14 Anthony Joonkyoo Yun Treatment of Seasonal Conditions Through Modulation of the Autonomic Nervous System
US9974909B2 (en) 2010-01-05 2018-05-22 Microdose Therapeutx, Inc. Inhalation device and method
US11229378B2 (en) 2011-07-11 2022-01-25 Otsuka Pharmaceutical Co., Ltd. Communication system with enhanced partial power source and method of manufacturing same
US11529071B2 (en) 2016-10-26 2022-12-20 Otsuka Pharmaceutical Co., Ltd. Methods for manufacturing capsules with ingestible event markers

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8912908B2 (en) 2005-04-28 2014-12-16 Proteus Digital Health, Inc. Communication system with remote activation
CN103259027A (en) 2005-04-28 2013-08-21 普罗透斯数字保健公司 Pharma-informatics system
EP2063771A1 (en) 2007-03-09 2009-06-03 Proteus Biomedical, Inc. In-body device having a deployable antenna
US8439033B2 (en) 2007-10-09 2013-05-14 Microdose Therapeutx, Inc. Inhalation device
US8540633B2 (en) 2008-08-13 2013-09-24 Proteus Digital Health, Inc. Identifier circuits for generating unique identifiable indicators and techniques for producing same
EP3906845A1 (en) 2009-04-28 2021-11-10 Otsuka Pharmaceutical Co., Ltd. Highly reliable ingestible event markers
SG184494A1 (en) 2010-04-07 2012-11-29 Proteus Biomedical Inc Miniature ingestible device
JP2014504902A (en) * 2010-11-22 2014-02-27 プロテウス デジタル ヘルス, インコーポレイテッド Ingestible device with medicinal product
US9756874B2 (en) 2011-07-11 2017-09-12 Proteus Digital Health, Inc. Masticable ingestible product and communication system therefor
WO2015112603A1 (en) 2014-01-21 2015-07-30 Proteus Digital Health, Inc. Masticable ingestible product and communication system therefor
AU2013293234B2 (en) 2012-07-23 2017-08-31 Otsuka Pharmaceutical Co., Ltd. Techniques for manufacturing ingestible event markers comprising an ingestible component
WO2014062674A1 (en) 2012-10-18 2014-04-24 Proteus Digital Health, Inc. Apparatus, system, and method to adaptively optimize power dissipation and broadcast power in a power source for a communication device
US11149123B2 (en) 2013-01-29 2021-10-19 Otsuka Pharmaceutical Co., Ltd. Highly-swellable polymeric films and compositions comprising the same
US10175376B2 (en) 2013-03-15 2019-01-08 Proteus Digital Health, Inc. Metal detector apparatus, system, and method
US9796576B2 (en) 2013-08-30 2017-10-24 Proteus Digital Health, Inc. Container with electronically controlled interlock
US10084880B2 (en) 2013-11-04 2018-09-25 Proteus Digital Health, Inc. Social media networking based on physiologic information
US11051543B2 (en) 2015-07-21 2021-07-06 Otsuka Pharmaceutical Co. Ltd. Alginate on adhesive bilayer laminate film
KR102215238B1 (en) 2016-07-22 2021-02-22 프로테우스 디지털 헬스, 인코포레이티드 Electromagnetic sensing and detection of ingestible event markers

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5699649A (en) 1996-07-02 1997-12-23 Abrams; Andrew L. Metering and packaging device for dry powders
US5714007A (en) 1995-06-06 1998-02-03 David Sarnoff Research Center, Inc. Apparatus for electrostatically depositing a medicament powder upon predefined regions of a substrate

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4808413A (en) * 1987-04-28 1989-02-28 E. R. Squibb & Sons, Inc. Pharmaceutical compositions in the form of beadlets and method
US4795644A (en) * 1987-08-03 1989-01-03 Merck & Co., Inc. Device for pH independent release of drugs through the Donnan-like influence of charged insoluble resins
FR2771010B1 (en) * 1997-11-19 2003-08-15 Adir USE OF A COMBINATION OF AN ANGIOTENSIN CONVERSION ENZYME INHIBITOR AND A DIURETIC FOR THE TREATMENT OF MICROCIRCULATORY DISORDERS
US5960609A (en) * 1998-06-12 1999-10-05 Microdose Technologies, Inc. Metering and packaging method and device for pharmaceuticals and drugs
US6428809B1 (en) * 1999-08-18 2002-08-06 Microdose Technologies, Inc. Metering and packaging of controlled release medication

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5714007A (en) 1995-06-06 1998-02-03 David Sarnoff Research Center, Inc. Apparatus for electrostatically depositing a medicament powder upon predefined regions of a substrate
US5699649A (en) 1996-07-02 1997-12-23 Abrams; Andrew L. Metering and packaging device for dry powders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1408907A4

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7404968B2 (en) 1999-08-18 2008-07-29 Microdose Technologies, Inc. Metering and packaging of controlled release medication
US7670612B2 (en) 2002-04-10 2010-03-02 Innercap Technologies, Inc. Multi-phase, multi-compartment capsular delivery apparatus and methods for using same
WO2004010978A1 (en) 2002-07-25 2004-02-05 Glaxo Group Limited Multicomponent pharmaceutical dosage form
EP1528916B1 (en) * 2002-07-25 2012-12-26 Capsugel Belgium NV Multicomponent pharmaceutical dosage form
US20100260669A1 (en) * 2004-05-13 2010-10-14 Anthony Joonkyoo Yun Treatment of Seasonal Conditions Through Modulation of the Autonomic Nervous System
WO2007047371A2 (en) * 2005-10-14 2007-04-26 Microdose Technologies, Inc. Pharmaceutical packaging of an oral dosage combination
WO2007047371A3 (en) * 2005-10-14 2007-12-06 Microdose Technologies Inc Pharmaceutical packaging of an oral dosage combination
US9974909B2 (en) 2010-01-05 2018-05-22 Microdose Therapeutx, Inc. Inhalation device and method
US10434267B2 (en) 2010-01-05 2019-10-08 Microdose Therapeutx, Inc. Inhalation device and method
US11229378B2 (en) 2011-07-11 2022-01-25 Otsuka Pharmaceutical Co., Ltd. Communication system with enhanced partial power source and method of manufacturing same
US11529071B2 (en) 2016-10-26 2022-12-20 Otsuka Pharmaceutical Co., Ltd. Methods for manufacturing capsules with ingestible event markers
US11793419B2 (en) 2016-10-26 2023-10-24 Otsuka Pharmaceutical Co., Ltd. Methods for manufacturing capsules with ingestible event markers

Also Published As

Publication number Publication date
ZA200402466B (en) 2006-07-26
CA2614902A1 (en) 2002-12-05
CA2448997A1 (en) 2002-12-05
JP2005514966A (en) 2005-05-26
NZ529696A (en) 2008-07-31
CA2614899A1 (en) 2002-12-05
WO2002096347A3 (en) 2003-03-27
BRPI0209720A2 (en) 2017-06-13
AU2002303840A1 (en) 2002-12-09
EP1408907A2 (en) 2004-04-21
EP1408907A4 (en) 2010-02-24

Similar Documents

Publication Publication Date Title
EP1408907A2 (en) Metering and packaging of controlled release medication
US20020142035A1 (en) Metering and packaging of controlled release medication
US20040156903A1 (en) Metering and packaging of controlled release medication
CA2668884C (en) Layered pharmaceutical formulations
HU206044B (en) Process for producing compositions with controlled release of dihydropyridine derivatives as active ingredient
Belgamwar et al. Pulsatile drug delivery system
WO2006040779A3 (en) Controlled release gastric floating matrix formulation containing imatinib
EP2061448B1 (en) Sustained release formulation of naltrexone
CN104780915A (en) Compositions comprising statins, biguanides and further agents for reducing cardiometabolic risk
CZ420997A3 (en) Layered tablet for controlled release of active compounds
CN115697304A (en) Multi-drug formulation for biodegradable subcutaneous reservoir device
AU770828B2 (en) Metering and packaging of controlled release medication
AU2004200545B9 (en) Metering and packaging of controlled release medication
KR20200111705A (en) Drug delivery system
Patel et al. Current status of technologies and devices for chronotherapeutic drug delivery systems

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 529696

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2002731902

Country of ref document: EP

Ref document number: 1856/CHENP/2003

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2448997

Country of ref document: CA

Ref document number: 2002592860

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 10479438

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2004/02466

Country of ref document: ZA

Ref document number: 200402466

Country of ref document: ZA

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 2002731902

Country of ref document: EP

ENPW Started to enter national phase and was withdrawn or failed for other reasons

Ref document number: PI0209720

Country of ref document: BR

Free format text: PEDIDO RETIRADO FACE A IMPOSSIBILIDADE DE ACEITACAO DA ENTRADA NA FASE NACIONAL POR TER SIDO INTEMPESTIVA. O PRAZO PARA ENTRADA NA FASE NACIONAL EXPIRAVA EM 31.01.2003( 20 MESES - BR DESIGNADO APENAS), ELEICAO NAO COMPROVADA, E A PRETENSA ENTRADA NA FASE NACIONAL SO OCORREU EM 27.11.2003.

ENP Entry into the national phase

Ref document number: PI0209720

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20031127