WO2006031856A2 - Biosynchronous transdermal drug delivery - Google Patents

Biosynchronous transdermal drug delivery Download PDF

Info

Publication number
WO2006031856A2
WO2006031856A2 PCT/US2005/032672 US2005032672W WO2006031856A2 WO 2006031856 A2 WO2006031856 A2 WO 2006031856A2 US 2005032672 W US2005032672 W US 2005032672W WO 2006031856 A2 WO2006031856 A2 WO 2006031856A2
Authority
WO
WIPO (PCT)
Prior art keywords
drug
animal
human
time
timing
Prior art date
Application number
PCT/US2005/032672
Other languages
French (fr)
Other versions
WO2006031856A3 (en
Inventor
Guy Dipierro
Steven Giannos
Original Assignee
Chrono Therapeutics, 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 Chrono Therapeutics, Inc. filed Critical Chrono Therapeutics, Inc.
Priority to CA2580329A priority Critical patent/CA2580329C/en
Priority to EP05796698.8A priority patent/EP1802258A4/en
Priority to JP2007531458A priority patent/JP5254616B2/en
Priority to AU2005284908A priority patent/AU2005284908B2/en
Publication of WO2006031856A2 publication Critical patent/WO2006031856A2/en
Publication of WO2006031856A3 publication Critical patent/WO2006031856A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • A61K9/703Transdermal patches and similar drug-containing composite devices, e.g. cataplasms characterised by shape or structure; Details concerning release liner or backing; Refillable patches; User-activated patches
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/04Nitro compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/465Nicotine; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • A61K9/0009Galenical forms characterised by the drug release technique; Application systems commanded by energy involving or responsive to electricity, magnetism or acoustic waves; Galenical aspects of sonophoresis, iontophoresis, electroporation or electroosmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0015Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin by using microneedles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M39/00Tubes, tube connectors, tube couplings, valves, access sites or the like, specially adapted for medical use
    • A61M39/22Valves or arrangement of valves
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M2037/0007Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin having means for enhancing the permeation of substances through the epidermis, e.g. using suction or depression, electric or magnetic fields, sound waves or chemical agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0015Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin by using microneedles
    • A61M2037/0023Drug applicators using microneedles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0015Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin by using microneedles
    • A61M2037/0061Methods for using microneedles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/02General characteristics of the apparatus characterised by a particular materials
    • A61M2205/0266Shape memory materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/33Controlling, regulating or measuring
    • A61M2205/3331Pressure; Flow
    • A61M2205/3337Controlling, regulating pressure or flow by means of a valve by-passing a pump
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/50General characteristics of the apparatus with microprocessors or computers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0092Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin using ultrasonic, sonic or infrasonic vibrations, e.g. phonophoresis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/18Applying electric currents by contact electrodes
    • A61N1/20Applying electric currents by contact electrodes continuous direct currents
    • A61N1/30Apparatus for iontophoresis, i.e. transfer of media in ionic state by an electromotoric force into the body, or cataphoresis

Definitions

  • This invention relates, in general, to controlled drug delivery methods and systems, and, more specifically, to systems and methods for biosynchronous transdermal drug delivery.
  • the invention further relates to the field of chronobiology in that the invention systems can be designed to modulate active agent delivery in accordance with biological rhythms. Drugs, pharmaceuticals, and other bioactive substances are delivered transdermally into a body in a manner that is synchronized with biological processes and/or biological rhythms so as to improve performance of the substance in the body.
  • the invention also relates to overcoming active agent tolerance, which may be experienced from continuous administration, improve patient compliance, and in some cases reducing the amount of drug needed per dose due to advantages of biosynchronization.
  • onset and symptoms of diseases such as asthma attacks, coronary infarction, angina pectoris, stroke and ventricular tachycardia are circadian phase dependent.
  • variations during the 24 h day in pharmacokinetics have been shown for cardiovascular active drugs (propranolol, nifedipine, verapamil, enalapril, isosorbide 5-mononitrate and digoxin), anti-asthmatics (theophylline and terbutaline), anticancer drugs, psychotropics, analgesics, local anesthetics and antibiotics, to mention but a few.
  • controlled release refers generally to delivery mechanisms that make an active ingredient available to the biological system of a host in a manner that supplies the drug according to a desired temporal pattern.
  • Controlled release drug delivery systems may be implemented using: a) instantaneous release systems; b) delayed release systems, and c) sustained release systems. In most cases, controlled release systems are designed to maintain a sustained plasma level of an active ingredient in a drug within a human or animal host over a period of time.
  • Instantaneous release refers to systems that make the active ingredient available immediately after administration to the biosystem of the host.
  • Instantaneous release systems include continuous or pulsed intravenous infusion or injections. Such systems provide a great deal of control because administration can be both instantaneously started and stopped and the delivery rate can be controlled with great precision.
  • the administration is undesirably invasive as they involve administration via a puncture needle or catheter.
  • Delayed release refers to systems in which the active ingredient made available to the host at some time after administration. Such systems include oral as well as injectable drugs in which the active ingredient is coated or en-capsulated with a substance that dissolves at a known rate so as to release the active ingredient after the delay. Unfortunately, it is often difficult to control the degradation of the coating or encapsulant after administration and the actual performance will vary from patient to patient.
  • Sustained Release generally refers to release of active ingredient such that the level of active ingredient available to the host is maintained at some level over a period of time.
  • sustained release systems are difficult to control and exhibit variability from patient to patient. Due to the adsorption through the gastrointestinal tract, drug concentrations rise quickly in the body when taking a pill, but the decrease is dependent on excretion and metabolism, which cannot be controlled. In addition, the adsorption through the gastrointestinal tract in many cases leads to considerable side effects (such as ulcers), and can severely damage the liver.
  • Transdermal therapeutic systems have been developed primarily for sustained release of drugs in situations where oral sustained release systems are inadequate.
  • drugs cannot be effectively administered orally because the active ingredients are destroyed or altered by the gastrointestinal system.
  • the drug may be physically or chemically incompatible with the coatings and/or chelating agents used to implement sustained release.
  • a transdermal delivery system may provide sustained release over a period of days or weeks whereas orally administered drugs may offer sustained performance over only a few hours.
  • a wide variety of active substances can be delivered through transdermal systems so long as the active substance can be provided in a form that can cross the skin barrier, see for example, U.S. Patent 6,638,528, which is incorporated herein by reference.
  • transdermal delivery systems are passive, taking the form of a patch that is attached to the skin by an adhesive.
  • the TTS includes a quantity of the active substance, along with a suitable carrier if need be, in a reservoir, matrix or in the adhesive itself. Once applied, the active ingredient diffuses through the skin at a rate determined by the concentration of the active substance and the diffusivity of the active substance.
  • a variety of physical and chemical processes at the skin/patch boundary affect the delivery rate and may eventually inhibit drug delivery altogether.
  • the original performance target for controlled drug delivery is to achieve a zero-order release rate of the drug, so that a constant efficacious drug concentration is maintained in the blood plasma.
  • chronobiology and chronopharmacology have demonstrated the importance of biological rhythms to the dosing of medications as well as determine the influence of a patient's circadian or other biological rhythms on drug efficacy and efficiency.
  • This research reveals that certain disease symptoms follow a daily pattern, with peak symptoms at certain times of the day. It has been widely acknowledged that hormones, neurotransmitters and other intra-body compounds are released in different amounts at different times of the day pursuant to daily patterns.
  • Active transdermal delivery systems have been developed to help regulate the delivery rate by providing mechanisms to improve drag delivery over time by "pumping" the active ingredient.
  • One such system (U.S. Patent 5,370,635), describes a system for delivering a medicament and dispensing it to an organism for a relatively long period of time, for example at least a few days.
  • the device can be adapted for positioning on the surface of the skin of a human or possibly an animal body in order to apply a medicament thereto from the outer side thereof.
  • Conventional transdermal systems circumvent the disadvantages of the adsorption through the gastrointestinal tract, but they do not optimize or tailor the dosing regiment to offset peak symptoms.
  • transdermal chronotherapy is a biphasic profile, in which the drug concentration changes from a high to a low level (or vice versa) over time.
  • the system can be physically applied or removed to alter the drug level, patient compliance with this procedure may be difficult, particularly during inconvenient hours.
  • the delivery system may utilize an external regulator, as described in Fallon et al. (U.S. Patent 5,352,456, 1994) which illustrates a device for drug administration through intact skin that provides an initial pulse in the flux of the drug through the skin followed by a substantially lower flux of drug through the skin.
  • Fallon et al. (U.S Patent 5,820,875, 1998) later describe a device for the administration of a drug through an area of intact skin over a period of time in which the flux of the drug through the skin varies temporally in a controlled manner.
  • the device is such that the skin flux of the drug varies in a controlled manner over the period of administration, typically from a high flux in the initial stage of administration to a lower flux in the later stage of administration.
  • Transdermal temporally controlled drug delivery systems proposed by Giannos et al. (U.S. Patent 6,068,853, 2000) coupled pH oscillators with membrane diffusion in order to generate a periodic release of a drug or active ingredient transdermally, without external power sources and/or electronic controllers.
  • the intent was to address chronotherapy with a pulsatile transdermal system.
  • the strategy was based on the observation that a drug may be rendered charged or uncharged relative to its pKa value. Since only the uncharged form of a
  • a periodic delivery profile may be obtained by oscillating the pH of the drug solution (see Giannos, S. A., "Pulsatile Delivery of Drugs and Topical Actives," in “Novel Topical Actives and Delivery Systems: Cosmetics, Dermatologicals and Transdermals”, Edited by John. J. Wille, Jr.: Blackwell Publishing, Oxford UK (In press)).
  • an orally administered drug for arthritis treatment has suggested a chronotherapeutic approach using a delay release system. The delay is scheduled to release the active ingredient at the beginning of an interleukin 6 cascade that is believed to cause early morning stiffness in rheumatoid arthritis patients.
  • By attempting to synchronize the drug delivery with a biological cycle it is believed that low doses may be used to achieve desired results.
  • this system does not overcome the limitations of delayed release systems described above.
  • An individual's failure to comply with a dosing regimen e.g. failure to take one or more doses of a drug or taking too many doses, will have an adverse impact upon the success of the regimen.
  • Individuals may fail to comply with their drug dosing regimen for a number of reasons.
  • drug dosing regimens such as every 4 hours, i. e. 8-12-4-8 involve a rigid dosing schedule that may be incompatible with an individual's personal schedule.
  • Such a rigid dosing schedule when combined with normal human traits such as forgetfulness or denial of a medical condition, as well as a busy life, represent substantial obstacles to compliance with a drug dosing regimen.
  • the present invention describes methods for treating diseases, addictions and disorders in humans. These methods involve synchronizing and tailoring the administration of drug compounds with the body's natural circadian rhythms, in order to counteract symptoms when they are likely to be at their worst, and are accomplished by using an automated and pre programmable transdermal drug administration system.
  • This system can also utilize a pump or pressurized reservoir, and/or a system for removing depleted carrier solution, or other modulated dispensing actuator, in conjunction with micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers.
  • these methods synchronize and tailor drug administration to the human body's circadian rhythms to deliver varying dosages at varying times. This ensures that peak drug concentrations are present in the bloodstream to offset peak disease and addiction symptoms arising from variances and fluctuation in the body's natural circadian rhythms. Further, these methods ensure that less of a drug is in the bloodstream when disease and addiction symptoms are at there lowest. This minimizes negative side effects, and increases efficacy of the dosing regimen.
  • Figure 1 shows an exemplary device useful for implementing the present invention.
  • Figure 2A - 2B illustrate comparative drug release profiles demonstrating operation of the present invention.
  • FIG. 3 is a schematic illustration of a drug delivery device in accordance with the present invention.
  • permeation through the skin may be assisted by using micro- fabricated structures commonly referred to as Micro-needles, heating devices, iontophoretic devices, or sonophoretic devices that are attached to this device.
  • FIG. 4 is a schematic illustration of an alternative drug delivery device in accordance with the present invention.
  • permeation through the skin may be assisted by using micro-fabricated structures commonly referred to as Micro-needles, heating devices, iontophoretic devices, or sonophoretic devices that are attached to this device.
  • Figure 5 shows an exemplary administration profile for a stimulant delivery system.
  • Figure 6 shows an exemplary administration profile for a nicotine delivery system.
  • Figure 7 shows an exemplary administration profile for a nitroglycerin delivery system tailored to treat variant angina attacks.
  • Figure 8 illustrates an exemplary administration profile for a nitroglycerin delivery system tailored to treat stress-induced angina attack.
  • Figure 9 illustrates an exemplary administration profile for an indomethacin delivery system tailored to arthritis.
  • Figure 10 illustrates an exemplary administration profile for a valdecoxib delivery system tailored to treat arthritis.
  • Figure 11 illustrates an exemplary administration profile for a tulobuterol delivery system tailored to treat asthma.
  • Figure 12 illustrates an exemplary administration profile for a clonidine delivery system tailored to treat hypertension.
  • Figure 13 illustrates an exemplary administration profile for a selegiline delivery system tailored to treat CNS degenerative disorders (Parkinson's Disease).
  • Figure 14 illustrates an exemplary administration profile for a selegiline delivery system tailored to treat Alzheimer's Disease and attention deficit disorder.
  • Figure 15 illustrates an exemplary administration profile for a methylphenidate delivery system tailored to treat ADD.
  • Figure 16 illustrates an exemplary administration profile for a selegiline delivery system tailored to treat depression.
  • Figure 17 illustrates an exemplary administration profile for an oxybutynin delivery system tailored to urinary incontinence.
  • Figure 18 illustrates an exemplary administration profile for a zolmitriptan delivery system tailored to treat migraine.
  • Figure 19 illustrates an exemplary administration profile for a miglitol delivery system tailored to treat diabetes.
  • Figure 20 illustrates an exemplary administration profile for a fentanyl delivery system tailored to treat pain.
  • FIG. 2 IA-C illustrates an exemplary administration profile for 5-fluorouracil, doxorubicin and cisplatin delivery system tailored to treat cancer.
  • Figure 22 illustrates an exemplary administration profile for a zidovudine delivery system tailored to treat AIDS.
  • Figure 23 illustrates an exemplary administration profile for a gabapentin delivery system tailored to epilepsy.
  • Figure 24 illustrates an exemplary administration profile for a triprolidine delivery system tailored to treat colds and flu.
  • Biological rhythms are periodic fluctuations in biological characteristics over time, which also include circadian as well as seasonal variations.
  • the reality of circadian rhythms in animals including humans is well known (Halberg et al. J. Exp. Ther. Oncol. 3 (5) 223-260 (2003), Redfern et al. Chronobiology International 11 (4) 253-265 (1994)).
  • Circadian (approximately 24-hour) rhythms include the production of biological molecules such as endorphins, gonadotropin releasing hormone (GnRH), Cortisol and adrenaline. These regulate the body's temperature and heart rate, changes in characteristics of blood, such as stickiness, and behaviors such as wakefulness, sleep and periods of activity.
  • ultradian which are cycles shorter than a day (for example, the milliseconds it takes for a neuron to fire, or a 90-minute sleep cycle)
  • infradian referring to cycles longer than 24 hours (for example monthly menstruation) • seasonal, such as seasonal affective disorder (SAD), which causes depression in susceptible people during the short days of winter.
  • SAD seasonal affective disorder
  • the present invention involves precisely timing the administration of drugs so that they reach peak levels in synchronization with times when symptoms are likely to be at their worst, or times at which the drugs are believed to be more effective in the body and/or better tolerated by the patient.
  • the present invention is described in terms of a particular example of a drug delivery system that provides automated and precise control over dosing, with single-dose capability, (once while people sleep) or capability to administer separate and varying-sized doses many times throughout a multiple day period.
  • the present invention also relates to the administration of different, distinct, drugs and dosages at different times of the day.
  • ChronoDoseTM a miniaturized, automated and programmable non-invasive drug delivery system
  • the system enables controlling of the amount of drug exposed to the skin in a controlled time dependent way according to a programmed administration schedule that implements a desired dosage profile.
  • the present invention enables one to precisely control and vary the time of drug release and the amount of each dose, pursuant to an easily set pre-programmed dosage profile.
  • Research demonstrates that for certain symptoms, conditions and diseases, drug effects can be optimized when administered in a defined (and often varying) dosage at predefined times. This is known as Chronopharmacology (Reinberg, A.
  • FIG. 1 (100) shown in Fig. 1 can be used for a variety of active compositions, and is small, fully automated and programmable.
  • This system consists of a reusable wristwatch-like device
  • a small, disposable, 'reservoir' (103), which is about the size of a quarter or 1/2 dollar coin in a particular example, or is cylindrical in shape, that the user can simply pop-in to place on the watch-like platform.
  • This reservoir lasts, for example, up to 72 hours, depending on the application. Shorter and longer reservoir lifetimes are contemplated.
  • the device is readily adapted to be worn on the forearm, ankle, or other convenient body location.
  • the replaceable reservoir can include a description of an administration schedule that can be used to manually or automatically program device (100) with an administration schedule.
  • a written schedule can be printed on or affixed to the reservoir (101) or electrically programmed using volatile or non- volatile memory. In this manner, a dosing profile can be prescribed and filled by a pharmacy in much the same manner as a conventional drug prescription is handled today.
  • An exemplary implementation (shown in Fig. 3) comprises a collapsible drug reservoir, an expandable waste reservoir, a micro-pump, electronics for automation, a display, and a highly permeable membrane. Further, a heating element or a gas or air blowing apparatus may be used to assist evaporation of liquids into the waste reservoir or the environment.
  • An exemplary system is described in a United Kingdom patent entitled Transdermal Drug Delivery and Method filed on September 13, 2004, Application Number PCT/IB2004/ 002947, which is incorporated herein by reference.
  • the drug reservoir will contain between about 0.4 ml and 4 ml of drug formulation.
  • a tiny, miniaturized pump is activated at pre-programmed times and releases a predefined amount of drug formulation into the drug chamber, where the formulation comes into contact with diffusion matrix.
  • This diffusional matrix is in intimate contact with a highly permeable membrane.
  • This membrane rests on the skin, and provides for even diffusion of the drug over the device's drug absorption surface area.
  • This membrane works effectively with, and can be coated with, an adhesive, hydrogel or polymer substance, which allows for rapid transport kinetics.
  • the remaining drug formulation is either removed or evaporated from the membrane area via a waste chamber containing a desiccant, containing a hydrophilic substance (hydrogel) or the device is taken off.
  • the above described device may use permeation enhancers whereby permeation through the skin is assisted, such as mechanical permeation enhancers that include micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers.
  • mechanical permeation enhancers that include micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers.
  • a pressurized drug reservoir is used which minimizes or eliminates need for a micropump.
  • Electronics control a valve that allows controlled quantities of the drug to be applied to the drug chamber where the formulation comes into contact with highly permeable membrane.
  • permeation enhancers whereby permeation through the skin is assisted, such as mechanical permeation enhancers that include micro- fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers.
  • transdermal patches for the delivery of pharmaceutical agents. See, for example, U.S. Patent 5,370,635, the disclosure of which is incorporated herein by reference.
  • patches may be constructed using a saturated media, pressurized reservoirs, or unpressurized reservoirs with micropumps for continuous, pulsatile, or on-demand delivery of an active material.
  • a pharmaceutically acceptable composition of an active material may be combined with either mechanical skin penetration enhancers including, but not limited to, micro-fabricated structures commonly referred to as Micro-needles, heat, iontophoresis, or sonophoresis, or a wide range of chemical permeation enhancers such as oleic acid, ethanol, amino acids, oleyl alcohol, long chain fatty acids, propylene glycol, polyethylene glycol, isopropanol, ethoxydiglycol, sodium xylene sulfonate, ethanol, N- methylpyrrolidone, laurocapram, alkanecarboxylic acids, dimethylsulfoxide, polar lipids, N- methyl-2-pyrrolidone, and the like, which increase the permeability of the skin to the active material and permit the active
  • mechanical skin penetration enhancers including, but not limited to, micro-fabricated structures commonly referred to as Micro-needles, heat, iontophoresis, or sonophoresis,
  • compositions may be combined with one or more agents including, but not limited to, alcohols, moisturizers, humectants, oils, emulsifiers, thickeners, thinners, surface-active agents, fragrances, preservatives, antioxidants, vitamins, or minerals.
  • agents including, but not limited to, alcohols, moisturizers, humectants, oils, emulsifiers, thickeners, thinners, surface-active agents, fragrances, preservatives, antioxidants, vitamins, or minerals.
  • Device-skin interface coupling media and/or control membranes include, but are not limited to, ethylcellulose, hydroxypropyl cellulose, poly (ethylene co-vinyl acetate), polyvinyl pyrrolidone, poly (ethylene oxide), poly (ethylene vinyl alcohol) and the like, to provide the composition in gel or hydrogel form, which may be dissolved in solvents such as water, methylene chloride or ethanol evaporated to the desired viscosity, and then applied to backing material to provide a patch.
  • the control membranes can be any of the conventional materials such as microporous polyethylene, polyethylene co-vinyl acetate (EVA copolymer), polyurethane and the like.
  • Chronopharmacokinetics is defined as the predictable changes observed in the plasma levels of drugs and in the parameters used to characterize the pharmacokinetics of a drug. Studies on animals and humans indicate that the C max , T ma ⁇ , AUC and half-life often vary as a function of the hour of administration of the drug. Table 1 presents a list of medications for which temporal changes in pharmacokinetics have been documented.
  • Chronopharmacology is critical to optimized dosing but is not being implemented because no automated transdermal system exists, and (ii) these drugs can be transdermally absorbed passively (i.e., without the need for external modulation or pre- treatment such as sonophoresis, iontophoresis, electroporesis, microneedles, etc. or other permeation enhancement.
  • Example substances include caffeine and ephedrine, and a variety of over-the-counter (OTC) and prescription stimulants (for treating fatigue, sleep disorders, attention deficit disorders and a variety of other conditions) in addition to herbal supplements, nicotine (for smoking cessation), nitroglycerin (for treating heart attack and strokes), fentanyl (for treating chronic pain), albutamol (for treating asthma), and selegiline (for treating depression, attention deficit disorder or Parkinson's disease).
  • OTC over-the-counter
  • prescription stimulants for treating fatigue, sleep disorders, attention deficit disorders and a variety of other conditions
  • herbal supplements for treating fatigue, sleep disorders, attention deficit disorders and a variety of other conditions
  • nicotine for smoking cessation
  • nitroglycerin for treating heart attack and strokes
  • fentanyl for treating chronic pain
  • albutamol for treating asthma
  • selegiline for treating depression, attention deficit disorder or Parkinson's disease
  • the present invention can preprogram the times and amount of each dosage by precisely controlling the amount of drug exposed to the skin during each dosing. This feature is advantageous when a drug is best administered during sleep, e.g., 1 to 2 hours before waking up.
  • the present invention precisely counteracts peak disease symptoms and increase patient compliance.
  • the present invention represents the first true non-invasive chronopharmacological drug delivery device. While current transdermal applications are restricted to the dosage profile shown in Figure 2a, the automated implementation of the present invention can be programmed for a variety of drug delivery patterns to achieve customized patient dosing regiments for optimal therapy (Figure 2b).
  • controlled transdermal release of an active material such as a drug
  • the term 'controlled' or 'sustained' release of an active material includes continuous or discontinuous, linear or non-linear release of the active material according to a programmed schedule.
  • advantages of controlled release are the convenience of a single application for the patient, avoidance of peaks and valleys in systemic concentration which can be associated with repeated injections, the potential to reduce the overall dosage of the active material, lower body stress, and the potential to enhance the pharmacological effects of the active material.
  • a lower, sustained dose can also prevent adverse affects that are occasionally observed with infusion therapy.
  • controlled release drug therapy can free the patient from repeated treatment or hospitalization, thus offering the patient greater flexibility and improving patient compliance.
  • a controlled release formulation of certain drugs also provides an opportunity to use the drug in a manner not previously exploited or considered.
  • the present invention is particularly advantageous when (i) known chronopharmacological information indicates that a drug's effects can be optimized when administered in a defined dosage at a predefined time or times, and/or (ii) patient compliance with the dosing regimen is greatly increased due to automation, i.e. doses are required at inopportune times, i.e. at night while sleeping.
  • the present invention may be used to treat, cure, prevent, control or alleviate a wide range of conditions and symptoms.
  • the drug delivery regimen of the present invention is administered to treat a condition selected from the group consisting of vitamin and/or mineral deficiency, Cancer, Addiction, Arthritis, Parkinson's Disease, Attention Deficit Disorder, Cardiovascular Disorder, Cold/Flu Symptoms, Pain, Childhood Bronchial Asthma, Peptic Ulcer, Post-operative Recuperation, and so forth as shown below.
  • a contemplated consumer product is the ArisePatchTM. Most people experience difficulty and discomfort when waking early in the morning. According to a 2002 National Sleep Foundation poll 49 % of US adults age 18-29 have trouble waking in the morning and 41 % of US adults age 30-64 have trouble waking in the morning. There are 165,000,000 adults in the US alone age 18-64; meaning approximately 74,250,000 US adults age 18-64 have trouble waking in the morning. Chronotherapeutic Rationale:
  • the ArisePatch implementation of the present invention allows individuals, while asleep, to have an over-the-counter (OTC) or prescription stimulant automatically ad ⁇ ministered during a 1 -2 hour pre-wake-up period.
  • Fig. 5 illustrates an exemplary stimulant administration profile showing a blood plasma level of ephedrine in nanograms per milliliter on the vertical axis, with time on the horizontal axis. Stimulant concentrations will reach peak levels immediately prior to having to wake. Immediately upon waking up the individual will be alert and feel well rested.
  • the ArisePatchTM will eliminate the typical discomfort or difficulty associated with getting up early. This functionality is attractive to employed people getting up for work to ensure punctuality, and just about anyone who wants to offset morning discomfort associated with a late night, jet lag, or sickness.
  • Nicotine replacement has been the most frequently used therapy to support smokers in their effort to quit. Smokers report that the craving for a cigarette is greatest immediately upon waking in the morning. The time elapsed between wakening and the first cigarette is the best indicator of addiction. For most smokers this time only a few minutes. Additionally, research has shown that nicotine transdermal delivery is influenced by chronopharmacokinetics. Nicotine patch design should compensate by decreasing the dose at night as well as increasing the dose in the morning and after meals (Gries et al., 1998).
  • Nicotine ChronoDoseTM system An exemplary product contemplated by the present invention is called Nicotine ChronoDoseTM system.
  • the system can begin to administer nicotine (or nicotine analogs or any other smoking cessation compound including but not limited to bupropion) automatically during a one-hour period immediately prior to waking. This will relieve the smoker's peak craving upon waking without causing nightmares and insomnia. We believe that this system clearly provides a superior method for smoking cessation.
  • a more advanced nicotine replacement system than that described above is worn for three days at a time and is programmed to release nicotine in a daily rhythmic pattern such as shown in Fig. 6 to offset peaks in a smoker's cravings.
  • Fig. 6 illustrates an exemplary nicotine administration profile showing a blood plasma level of nicotine in nanograms per milliliter on the vertical axis, with time on the horizontal axis.
  • This implementation will reduce nicotine dependency by administering pre-programmed levels of nicotine pursuant to typical smoking patterns. For instance many smokers report that cravings for a cigarette are greatest upon waking up, after lunch, mid afternoon, after dinner and before bedtime.
  • This implementation of the present invention will automatically release larger doses of nicotine to offset peak cravings and no nicotine when cravings are typically at a minimum.
  • the present invention may be delivered in a preprogrammed manner for each treatment regimen. The only involvement by the user will be the replacement of the 'reservoir' every three days, and the replacement of the platform housing as needed.
  • This implementation represents a tremendous move forward in nicotine replacement therapy, and is far superior to the old-technology systems that simply release the same amount of nicotine all day and night.
  • the present invention one can systematically decrease a smoker's tolerance without increasing dependence (the result of a constant flow) and better wean a smoker off nicotine. This will allow the smoker to better 'tailor-down' and decrease the amount of nicotine he needs to quit. Modern smoking cessation is much more than nicotine replacement therapy.
  • Programs also include weight control, diet and psychological support. The present invention fits well into these programs, since it addresses the key component of being able to quit smoking by efficiently countering the withdrawal symptoms while doing away with the negative side effects of current nicotine replacement therapy systems, namely sleep disturbance.
  • Nitroglycerin effectively combats angina attacks, if administered in optimal doses.
  • nitroglycerine loses its effectiveness and requires higher and higher dosages when administered constantly. Our bodies become tolerant to it. Current systems cannot stop or decrease the release of nitroglycerine when disease symptoms are lowest. Thus, these current 'dumb' patches cannot offset the critical angina phase by releasing more of the drug, nor can they shut down or stop nitroglycerin administration when the body doesn't need it. It is a 'one dose fits all' type of scenario once each "dumb" patch is applied to the patient. Chronotherapeutic Rationale:
  • the method in accordance with the present invention utilizes an automated transdermal system in order to transdermally administer more nitroglycerin during the critical angina phase to ensure adequate offset of these symptoms and less nitroglycerin when it is not needed so that no tolerance builds up.
  • Our method utilizes a 'smart' patch medicine system at this time to offset these peak critical phases in the disease cycle arising due to the human body's circadian rhythm.
  • the pre-programmable automated transdermal system is worn around the wrist - like a watch (or the forearm arm or ankle) and releases nitroglycerin in optimal dosages at times that are optimally synchronized. This is pursuant to a pre-programmed and tailored dosage profile.
  • Current nitroglycerin patches only have the capability to release a constant dose of nitroglycerin over a period of time.
  • Current nitroglycerin patches simply cannot alter or vary dosages to increase dosages at different times of the day, and decrease dosages at other times of the day.
  • the nitroglycerin system in accordance with the present invention has three primary advantages over current nitroglycerin patches.
  • Current nitroglycerin patches have release rates that stay constant and do not increase to offset critical phases, and do not decrease as symptoms decrease.
  • Second, our system solves the tolerance issue by releasing less (or no) nitroglycerin in off-peak hours, and then releasing nitroglycerin at just the right time so that it is present during critical periods, without increasing tolerance.
  • the nitroglycerin system represents an ideal delivery system for patients who use nitroglycerin regularly for the treatment and/or the prevention of heart attacks and strokes.
  • Patient compliance regarding the timing and dose of heart attack medication is crucial.
  • Patient non-compliance with physician's instructions for this is often a cause of re- hospitalization, according to the US Department of Health and Human Services.
  • the system solves this problem, and will decrease the need for re-hospitalization by dramatically increasing patient compliance.
  • This system can be either an 'wear each night and remove in the morning' system, whereby it only releases nitroglycerin automatically to offset the critical angina phase in the morning, or a 'total solution' system, that is worn for a period of 24 hours to several days, and that administers nitroglycerin in tailored amounts and at tailored times as synchronized with the body's circadian rhythm (and conveniently taken off while showering or swimming).
  • the system is an innovative new drug therapy for angina.
  • the system in accordance with the present invention ensures that nitroglycerin will circulate in the bloodstream exactly when the patient needs it, and without any build up tolerance.
  • the present invention is superior to current steady release nitroglycerin patches.
  • Our system's increased advantages are extremely relevant for those patients with moderate to severe angina.
  • Fig. 7 shows an exemplary administration profile for a nitroglycerin delivery system tailored to treat variant angina attacks or angina pectoris.
  • This type of angina attack has a peak frequency in many patients between the hours of 2:00 and 4:00 AM. This is a particularly difficult time to wake up to take a drug such as nitroglycerin.
  • an administration profile substantially like that shown in Fig. 7 is automatically administered.
  • the vertical axis indicates blood plasma level in nanograms per milliliter, and the horizontal axis indicates time from 10:00 PM through the night to 8:00AM.
  • Fig. 8 illustrates an exemplary administration profile for a nitroglycerin delivery system tailored to treat stress-induced angina attack.
  • the vertical axis indicates blood plasma level in nanograms per milliliter, and the horizontal axis indicates time from 12:00 AM through the day until about 4:00 PM.
  • the administration profile shown in Fig. 8 provides a high blood plasma concentration throughout the waking hours of a day when stress is likely occur.
  • APPLICATIONS Arthritis (Examples: Indomethacin, Valdecoxib) An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automatically in the morning, just before a person awakes and the symptoms of arthritis are the worst. Later, towards mid-day, the drug concentration is also increased. Then in the evening, the drug dose is increased prior to bedtime.
  • osteoarthritis and rheumatoid arthritis both show distinctive circadian patterns of pain. While many people feel stiff for an hour or so after first getting up in the morning, people with osteoarthritis typically hurt most and have the most difficulty moving in the afternoon and evening. Those with rheumatoid arthritis almost always feel much worst in the morning. By dosing at night, early morning and mid-day, the benefits of non-steroidal anti-inflammatory drugs (NSAIDs) and cyclocoygenase-2 inhibitors (COX-2) can be maximized and side effects reduced. Examples of medications for arthritis include:
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
  • BPC should be in the highest therapeutic range of between 0.5-2.0 mcg/ml. Peak 2 (Medium)
  • BPC should be in the medium therapeutic range of between 0.25 - 1.5 mcg/ml.
  • BPC should be in the highest therapeutic range of between 0.5 to 2.0 mcg/ml.
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest) 5:00am-9:00am: BPC should be in the highest therapeutic range of between 50-175 ng/ml. Peak 2 (Medium)
  • BPC should be in the medium therapeutic range of between 21-125 ng/ml.
  • BPC should be in the highest therapeutic range of between 50 to 175 ng/ml.
  • the automated transdermal asthma system automatically administers a morning dose of albuterol, tulobuterol, salmeterol, beta 2 agonist or any other antiarrhythmic drug (an 'Asthma drug') to combat the peak symptom of morning asthma attacks known as the 'morning dip'.
  • Asthma attacks occur 100 (one hundred) times more often between the hours 4 A.M. and 6 A.M., when most people are asleep. This is due to the early morning deterioration of respiratory function known as 'morning dip', which is the time of day that respiratory function is at its lowest. These early morning asthma attacks cause great distress to sufferers and care providers.
  • the morning dip represents the dip in respiratory function at this time when asthma attacks are 100 times more likely to occur.
  • Our system effectively combats the morning dip by releasing more Asthma drug at this time to offset this peak morning symptom.
  • our 'smart' patch varies the level of drug in the bloodstream so that drug concentrations are highest when respiratory function is at its lowest.
  • the Asthma system has two primary advantages over current patches.
  • First, the system of the present invention utilizes its core competitive advantage to automatically and precisely release tulobuterol or other asthma drugs in peak amounts to offset the peak symptoms associated with the morning dip.
  • Current patches have release rates that stay constant and do not increase to offset this peak critical phases, and do not decrease as symptoms decrease.
  • Second, our system accomplishes 1 and 2 above automatically, without the need for a patient to wake up to take a drug at this critical phase, which does away with the need for any increased patient compliance.
  • the automated transdermal system for asthma is worn around the wrist like a watch (or the forearm arm or ankle) and releases albuterol or other asthma drugs in optimal dosages at times that are optimally synchronized, especially to offset the morning dip, pursuant to a pre-programmed and tailored dosage profile.
  • Current Asthma patches only have the capability to release a constant dose over a period of time.
  • Current Asthma patches simply cannot alter or vary dosages to increase dosages at different times of the day, and decrease dosages at other times of the day.
  • the system is an innovative new drug therapy for asthma. With its superior advantage of optimized and automated time and dose administration synchronized with our circadian rhythms, our system ensures that tulobuterol or another asthma drug will circulate in increased amounts in the bloodstream exactly when the patient needs it. For these reasons, our system is superior to current steady release patches. Our system's increased advantages are extremely relevant for those patients with moderate to severe asthma.
  • the time/dose chart should appear as shown in Figure 11
  • the automated transdermal system for hypertension has two primary advantages over current patches.
  • First, our system utilizes its core competitive advantage to automatically and precisely release clonidine or other hypertension drugs in peak amounts to offset the peak symptoms associated with the dangerous morning symptoms.
  • Current hypertension patches have release rates that stay constant and do not increase to offset this peak critical phases, and do not decrease as symptoms decrease.
  • Second, our system accomplishes 1 and 2 above automatically, without the need for a patient to wake up to take a drug at this critical phase, which does away with the need for any increased patient compliance.
  • the clonidine automated transdermal system utilizes clonidine, (or another hypertension drug) an effective drug that combats high blood pressure.
  • the clonidine automated transdermal drug delivery system has an automated morning release of Clonidine to combat the peak symptom of morning heart attacks. Blood pressure differs at different times of the day. Blood pressure surges upon waking, and is lower by 20 to 30 per cent while sleeping.
  • Our preprogrammed automatic transdermal system utilizes its core competitive advantage by releasing clonidine in a tailored fashion to counter high blood pressure when symptoms are highest, while releasing less clonidine when symptoms are less severe.
  • the time/dose chart should appear as shown in Figure 12
  • the selegiline automated transdermal drug delivery system gives an automated morning release of selegiline to combat the peak symptom of morning depression without the side effect of sleep disturbances.
  • the system in accordance with the present invention is applied before bed. It does not release the drug until one or two hours before morning, so symptom of morning depression would be corrected by our system without subjecting the patient to sleep disturbances.
  • the time/dose chart should appear as shown in Figure 13
  • Selegiline is an effective MAO inhibitor for the treatment of depression, Alzheimer's and Attention Deficit Disorder.
  • oral selegiline produces many undesirable side effects.
  • a transdermal form of selegiline, EMSAMTM is currently being developed. However, it also produces sleep disturbances as well. It is believed that the system in accordance with the present invention would be superior to conventional selegiline product delivery systems.
  • the selegiline automated transdermal drug delivery system gives an automated morning release of selegiline to combat the peak symptom of morning depression without the side effect of sleep disturbances.
  • the system in accordance with the present invention is applied before bed. It does not release the drug until one or two hours before morning, so symptom of morning depression would be corrected by our system without subjecting the patient to sleep disturbances.
  • Ritalin is indicated as an integral part of a total treatment program which typically includes other remedial measures (psychological, educational, social) for a stabilizing effect in children with a behavioral syndrome characterized by the following group of developmentally inappropriate symptoms: moderate-to-severe distractibility, short attention span, hyperactivity, emotional lability, and impulsivity.
  • Methylphenidate is usually administered in divided doses 2 or 3 times daily, preferably 30 to 45 minutes before meals. Patients who are unable to sleep if medication is taken late in the day should take the last dose before 6 p.m. Since the suggested first dose is early in the morning, it would be beneficial to automatically control the dosage.
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
  • BPC should be in the highest therapeutic range of between 8-25 ng/ml. Peak 2 (Highest)
  • BPC should be in the highest therapeutic range of between 8-25 ng/ml.
  • BPC should be in the highest therapeutic range of between 8 to 25 ng/ml.
  • the time/dose chart should appear as shown in Figure 15
  • Selegiline is an effective MAO inhibitor for the treatment of depression, Alzheimer's and Attention Deficit Disorder.
  • oral selegiline produces many undesirable side effects.
  • a transdermal form of selegiline, EMSAMTM is currently being developed. However, it also produces sleep disturbances as well. It is believed that the system in accordance with the present invention would be superior to conventional selegiline product delivery systems.
  • the selegiline automated transdermal drug delivery system gives an automated morning release of selegiline to combat the peak symptom of morning depression without the side effect of sleep disturbances.
  • the system in accordance with the present invention is applied before bed. It does not release the drug until one or two hours before morning, so symptom of morning depression would be corrected by our system without subjecting the patient to sleep disturbances.
  • the time/dose chart should appear as shown in Figure 16
  • An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automatically at night while asleep, and to decrease concentrations during the daytime work hours, and again to slightly increase drug concentrations after work and prior to bed.
  • Oxybutynin The primary adverse side effect of Oxybutynin is daytime sleepiness, daytime attention and cognitive deficits, drowsiness, dizzyness, blurred vision, (must use caution when driving, operating machinery, or performing other hazardous activities). Therefore, it seems that a dose in the lower end of the therapeutic range should be administered during the daytime, with a slightly larger dose administered after working hours, and with an even higher dose administered during the sleeping hours.
  • This dosing regimen would also give the user a higher dose at night, when one sleeps. At this time, increased drowsiness would be advantageous as well as providing a period of undisturbed sleep due to the inhibition of urge incontinence.
  • Medications for incontinence include:
  • the mean maximum blood plasma concentration following oral dosing with 5 mg oxybutynin or transdermally with 39 mg is 3 ng/mL. Blood plasma concentration between 1 and 3 ng/ml
  • Theoretical unenhanced transdermal flux for oxybutynin (Berner-Cooper predictive model) is 10.98 ug/cm 2 /hr.
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
  • BPC should be in the highest therapeutic range of between 2.5-4.5 ng/ml. Peak 2 (Low)
  • BPC should be in the lowest therapeutic range of between 0.75 - 1.5 ng/ml.
  • BPC should be in the medium therapeutic range of between 1.5 to 2.5 ng/ml.
  • the time/dose chart should appear as shown in Figure 17
  • An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automaticaly in the evening to provide needed medication, in the very early morning (0200 - 0400) while asleep, and again later on (0800 - 1000) upon waking. Then, during the daytime work hours, decrease concentrations to allow for normal activities.
  • Migraine, cluster and tension-type headaches may produce a headache that awakens an individual in the early morning hours (usually between 2 and 4 AM), or is present upon awakening. Those individuals with chronic tension-type headache are most likely to be awakened in the early morning hours due to headache. This headache also tends to be at its worst severity at that time of day. A variety of causes may account for this early-morning pattern to the headaches.
  • Medications for headache and migraine include: Abortive Medications
  • Analgesics with caffeine such as Excedrin® Migraine (acetaminophen, aspirin and caffeine).
  • Analgesics with caffeine and barbiturates such as Fiorinal® (butalbital, aspirin and caffeine) and Fioricet®(butalbital, acetaminophen and caffeine).
  • Non steroidal antiinflammatory drugs such as Advil® (ibuprofen), and Aleve® (naproxen sodium).
  • Ergotamines such as cafergot® (caffeine and ergotamine tartrate) and Migranal® (dihydroergotamine).
  • Triptans such as Zomig®(zolmitriptan), Maxalt® (rizatriptan), Imitrex® (sumatriptan), Frova® (frovatriptan), Axert® (almotriptan) and Amerge® (naratriptan).
  • Excedrin Migraine is a registered trademark of Bristol-Myers Squibb Company
  • Fiorinal and Fioricet are registered trademarks of Novartis Pharmaceuticals Corporation
  • Advil is a registered trademark of Whitehall-Robbins Healthcare
  • Maxalt is a registered trademark of Merck & Co., Inc.
  • Imitrex is a registered trademark of GlaxoSmithKline
  • Frova is a registered trademark of Elan Pharmaceuticals/UCB Pharma, Inc.
  • Axert is a registered trademark of Pharmacia
  • Beta blockers such as Inderal® (propranolol)*, Blocadren ® (timolol maleate)*, and metoprolol.
  • Calcium-channel blockers such as Cardizem® (diltiazem) and Procardia® (nifedipine).
  • Antidepressants such as Prozac® (fluoxetine), Paxil® (paroxetine) and Zoloft® (sertraline).
  • Anticonvulsants such as Depakote® (valproic acid or divalproex sodium).
  • NSAIDs such as Orudis® (ketoprofen) and Aleve® (naproxen sodium).
  • Inderal is a registered trademark of AstraZeneca
  • Blocadren is a registered trademark of Merck & Co, Inc.
  • Procardia is a registered trademark of Pfizer Inc.
  • Prozac is a registered trademark of Eli Lilly and Company
  • Paxil is a registered trademark of GlaxoSmithKline
  • Zoloft is a registered trademark of Pfizer Inc.
  • Depakote is a registered trademark of Abbott Laboratories
  • Orudis is a registered trademark of Aventis Pharmaceuticals
  • Theoretical unenhanced transdermal flux for zolmitriptan (Berner-Cooper predictive model) is 6.02 ug/cm 2 /hr.
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times:
  • BPC should be in the highest therapeutic range of between 3.5-4.0 ng/ml.
  • BPC should be in the highest therapeutic range of between 3.5-4.0 ng/ml.
  • BPC should be in the lowest therapeutic range of between 1.0- 3.0 ng/ml.
  • An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automaticaly in the morning (0800), midday (1200) and evening (1800) which coincide with mealtimes.
  • Miglitol is indicated as an adjunct to diet to improve glycemic control in patients with non-insulin-dependent diabetes mellitus (NIDDM) whose hyperglycemia cannot be managed with diet alone.
  • NIDDM non-insulin-dependent diabetes mellitus
  • Theoretical unenhanced transdermal flux for miglitol (Berner-Cooper predictive model) is 49.24 ug/cm 2 /hr.
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
  • BPC should be in the lowest therapeutic range.
  • pain induced experimentally was reported to be maximal in the morning, or in the afternoon or at night.
  • a circadian pattern of pain has been seen in patients suffering from pain produced by different diseases. For instance, highest toothache intensity occurred in the morning, while biliary colic, migraine, and intractable pain were highest at night.
  • Patients with rheumatoid anhritis reported peak pain early in the morning, while those with osteoarthritis of the knee indicated that the maximal pain occurred at the end of the day.
  • the effectiveness of opioids appears also to vary according to time of day, but large differences in the time of peak and low effects were found. Peak pain intensity and narcotic demands occur early in the morning, or it can be at the end of the day. Pain is a complex phenomenon and specific to each clinical situation.
  • An automated, and programmed, pulsatile transdermal drug delivery regimen is needed to substantially increase blood plasma concentrations of Fentanyl or other pain medications, automatically between 3:00am and 8:00 am, while people sleep, where pain results from inflammation, because cortisone, a key inflammation fighter, is lowest in the body at that time. Additionally, an automated, and programmed, pulsatile transdermal drug delivery regimen is needed to substantially increase blood plasma concentrations of Fentanyl or other pain medications automatically between 6:00am and 9:00am for Ankylosing spondylitis pain, and in mid-afternoon for Osteoarthritis pain.
  • Other pain medication includes: codeine, dihydrocodeine, hydrocodone or hydromorphone, Sufentanil, Nalbuphine, Buprenorphine, Hydromorphone and any type of opiate derivative.
  • transdermal pain management since they are effective, there is considerable hepatic first pass effect and a short half life, and they are skin permeable.
  • BPC should be in a moderate therapeutic range of between l-3ng/m. Peak 3 (Middle)
  • BPC should be in the lowest therapeutic range of between 2- 5g/ml.
  • Circadian fluorodeoxyuridine (FUDR) continuous infusion (70% of the daily dose given between 15:00 h and 21 :00 h) has been shown effective for metastatic renal cell carcinoma resulting in 29% objective response and stable disease of more than 1 yr duration in the majority of patients. Toxicity is reduced markedly when FUDR infusion is modulated to circadian rhythms
  • Chronotherapy has also been used to lower the amount of side effects from chemotherapy drugs. Over the years, doctors have realized that by giving two of these drugs, Adriamycin and cisplatin, in the morning and evening, respectively, side effects could be cut in half.
  • pulsatile delivery should have blood plasma concentrations (BPC) as set forth for each specific medication.
  • Adherence to antiretroviral therapy for the treatment of HIV infection and AIDS has become one of the most important clinical challenges among HIV health care providers and patients. Adherence to the prescribed regimen may predict which patients achieve undetectable viral loads. Unfortunately, non-adherence is common in antiretroviral therapy and has been associated with increases in viral load and the development of drug resistance. Efforts to maximize patient adherence are critical for suppressing HIV replication and preventing the transmission of drug-resistant virus.
  • BPC should be in the highestest therapeutic range.
  • Theoretical unenhanced transdermal flux for zidovudine (Berner-Cooper predictive model) is 17.94 ug/cm 2 /hr.
  • anti-seizure medications also called anticonvulsant or anti-epilepsy drugs
  • Some are specifically designed not to interfere with the activity of other drugs, including birth control pills. They include gabapentin (Neurontin), lamotrigme (Lamictal), topiramate (Topamax), tiagabine (Gabatril), levetiracetam (Keppra), and oxcarbazepine (Trileptal).
  • BPC should be in the highestest therapeutic range.
  • Cold and flu symptoms are worst from midnight until the early morning because the concentration of Cortisol is lowest at that time.
  • Current nighttime cold and flu medication end up losing efficacy by early morning when cold and flu symptoms are highest. Therefore people suffering from a cold or flu are often unpleasantly awoken by an increase in symptoms, cutting sleep short.
  • the present invention will automatically deliver a larger dose of medication and immuno-boosters in the early morning hours to more effectively combat the peak cold and flu symptoms that occur in the morning.
  • This implementation uses prescription or OTC cold medicine alone or optionally in combination with certain transdermally efficacious vitamins and immune system boosters to provide a total solution to cold and flu ailments.
  • This is the first cold therapy that combines OTC medicine with supplemental immuno-boosters in a comprehensive and automated manner.
  • the Cold and Flu automated transdermal drug delivery system utilizes OTC cold medicine, Vitamin C, Echinacea, and Zinc to provide a total solution to cold and flu ailments, and all while a person sleeps.
  • the Cold/Flu system releases these combination of compounds every 2 hours throughout the night, with a higher dosage of compounds being released in the morning to combat these proven middle of the night and early morning symptoms, which are the worst of the day. Users will experience less severe cold and flu symptoms during the morning hours, will not have their sleep cycle cut short, and will wake up feeling symptom-free.
  • Vitamin and Herbal Supplementation In yet another application, a series of weight loss vitamins and supplements is administered in small distinct doses many times over several days. Vitamins and supplements are absorbed by the body in small dosages. Contrary to popular belief, once-a- day products are not maximally effective because excess dosages are excreted unused.
  • This implementation of the present invention precisely controls the timing and dosage of small but distinct amounts of vitamins and supplements during a 24-hour period to ensure that vitamins and supplements are constantly bio-available for optimal absorption and cellular function. Greater doses are automatically released prior to mealtimes to counter appetite cravings, resulting in a much more effective diet program.
  • the present invention is particularly useful in applications in which it is necessary and/or desirable to start the administration of a drug, stop the administration of a drug, and/or increase/decrease the dosage of a drug at a time when it is inconvenient or impossible for a patient to initiate the necessary actions.
  • This is particularly useful for a wide variety of drug administration applications that benefit when an administration is started, stopped, or changed while a person is sleeping.
  • chronotherapy As research and knowledge of chronotherapy increases, it is contemplated that a wide variety of applications will be discovered in which benefit is realized by starting, stopping and/or changing the drug administration while a patient sleeps.
  • treatment is continued as needed to provide superior symptomatic relief, prevent exacerbation of symptoms, and/or prevent and/or delay progression of the disease state or condition in the patient, or until it is no longer well tolerated by the patient, or until a physician terminates treatment.
  • a physician may monitor one or more symptoms and/or serum levels of active material and/or metabolic by-product(s) in a patient being treated according to this invention and, upon observing attenuation of one or more symptoms for a period of time, conclude that the patient can sustain the positive effects of the above-described treatment without further administration for a period of time. When necessary, the patient may then return at a later point in time for additional treatment as needed.
  • 'day' means a 24-hour period.
  • 'for at least three consecutive days' means for at least a 72-hour period.
  • a physician may monitor one or more symptoms and/or serum levels in the patient and, upon observing an improvement in one or more of the parameters for a period of time, conclude that the patient can sustain the positive effects of the treatment without further administration of the active material for a period of time.
  • the active material is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition.
  • a pharmaceutical composition comprising an active material in association with a pharmaceutically acceptable diluting substance or carrier, wherein the active material is present in an amount for effective treating or preventing a particular condition.
  • kits for treating a particular symptom, condition and/or disease and/or improving a particular biological function comprising one or more containers comprising one or more active compositions in accordance with this invention.
  • Such kits can also include additional drugs or therapeutics for co-use with the active composition for treatment or prevention of a particular symptom, condition and/or disease and/or improving a particular biological function.
  • the active composition and the drug can be formulated in admixture in one container, or can be contained in separate containers for simultaneous or separate administration.
  • the kit can further comprise a device(s) for ad-ministering the compounds and/or compositions, such as device 100 shown in Fig.

Abstract

A delivery mechanism and device for the passive periodic release of a drug or an active ingredient and the methods for synchronizing the administration of compounds with the human body's natural circadian rhythms and addiction rhythms. This strategy is intended to counteract disease states and symptoms when they are likely to be at their worst by using an automated and pre-programmable transdermal or other drug administration system.

Description

BIOSYNCHRONOUS TRANSDERMAL DRUG DELIVERY Field of the Invention
This invention relates, in general, to controlled drug delivery methods and systems, and, more specifically, to systems and methods for biosynchronous transdermal drug delivery. The invention further relates to the field of chronobiology in that the invention systems can be designed to modulate active agent delivery in accordance with biological rhythms. Drugs, pharmaceuticals, and other bioactive substances are delivered transdermally into a body in a manner that is synchronized with biological processes and/or biological rhythms so as to improve performance of the substance in the body. The invention also relates to overcoming active agent tolerance, which may be experienced from continuous administration, improve patient compliance, and in some cases reducing the amount of drug needed per dose due to advantages of biosynchronization.
Background of the invention
In the field of drug delivery, it is recognized that supplying the drug in a correct temporal pattern is an important attribute of any drug delivery methodology. Controlled release drug delivery systems are intended to improve the response to a drug and/or lessen side effects of a drug. The recurring interest in chronopharmacology demonstrates the fact that biological rhythms are an important aspect of clinical pharmacology and should be taken into account when evaluating drug delivery systems (Hrushesky, W., J. Cont. ReI. 19:363 (1992), Lemmer, B., Adv. Drug Del. Rev. 6:19 (1991), Youn, C. B. J. Cont. ReI. 98 (3) 337 (2004) and Youn, C. B. J., Ed., "Chronopharmaceutics," John Wiley & Sons, New York (In preparation)).
The onset and symptoms of diseases such as asthma attacks, coronary infarction, angina pectoris, stroke and ventricular tachycardia are circadian phase dependent. In humans, variations during the 24 h day in pharmacokinetics (chrono-pharmacokinetics) have been shown for cardiovascular active drugs (propranolol, nifedipine, verapamil, enalapril, isosorbide 5-mononitrate and digoxin), anti-asthmatics (theophylline and terbutaline), anticancer drugs, psychotropics, analgesics, local anesthetics and antibiotics, to mention but a few. Even more drugs have been shown to display significant variations in their effects throughout the day (chronopharmacodynamics and chronotoxicology) even after chronic application or constant infusion (Ohdo, S. Drug Safety 26 (14) 999-1010 (2003)). Moreover, there is clear evidence that dose/concentration-response relationships can be significantly modified based on the time of day. Thus, circadian time has to be taken into account as an important variable influencing a drug's pharmacokinetics and its effects or side-effects (Bruguerolle, B., Clin. Pharmacokinet. Aug. 35 (2) 83-94 (1998)).
Studies indicate that the onset of certain diseases show strong circadian temporal dependency. This has led to the need for timed patterning of drug delivery as opposed to constant drug release (Lemmer B., Ciba Found Symp. 183:235-47; discussion 247-53 (1995).
The term "controlled release" refers generally to delivery mechanisms that make an active ingredient available to the biological system of a host in a manner that supplies the drug according to a desired temporal pattern. Controlled release drug delivery systems may be implemented using: a) instantaneous release systems; b) delayed release systems, and c) sustained release systems. In most cases, controlled release systems are designed to maintain a sustained plasma level of an active ingredient in a drug within a human or animal host over a period of time.
Instantaneous release refers to systems that make the active ingredient available immediately after administration to the biosystem of the host. Instantaneous release systems include continuous or pulsed intravenous infusion or injections. Such systems provide a great deal of control because administration can be both instantaneously started and stopped and the delivery rate can be controlled with great precision. However, the administration is undesirably invasive as they involve administration via a puncture needle or catheter.
Delayed release refers to systems in which the active ingredient made available to the host at some time after administration. Such systems include oral as well as injectable drugs in which the active ingredient is coated or en-capsulated with a substance that dissolves at a known rate so as to release the active ingredient after the delay. Unfortunately, it is often difficult to control the degradation of the coating or encapsulant after administration and the actual performance will vary from patient to patient.
Sustained Release generally refers to release of active ingredient such that the level of active ingredient available to the host is maintained at some level over a period of time. Like delayed release systems, sustained release systems are difficult to control and exhibit variability from patient to patient. Due to the adsorption through the gastrointestinal tract, drug concentrations rise quickly in the body when taking a pill, but the decrease is dependent on excretion and metabolism, which cannot be controlled. In addition, the adsorption through the gastrointestinal tract in many cases leads to considerable side effects (such as ulcers), and can severely damage the liver.
Transdermal therapeutic systems (TTS) have been developed primarily for sustained release of drugs in situations where oral sustained release systems are inadequate. In some cases, drugs cannot be effectively administered orally because the active ingredients are destroyed or altered by the gastrointestinal system. In other cases the drug may be physically or chemically incompatible with the coatings and/or chelating agents used to implement sustained release. In other cases a transdermal delivery system may provide sustained release over a period of days or weeks whereas orally administered drugs may offer sustained performance over only a few hours. A wide variety of active substances can be delivered through transdermal systems so long as the active substance can be provided in a form that can cross the skin barrier, see for example, U.S. Patent 6,638,528, which is incorporated herein by reference.
In most cases transdermal delivery systems are passive, taking the form of a patch that is attached to the skin by an adhesive. The TTS includes a quantity of the active substance, along with a suitable carrier if need be, in a reservoir, matrix or in the adhesive itself. Once applied, the active ingredient diffuses through the skin at a rate determined by the concentration of the active substance and the diffusivity of the active substance. However, a variety of physical and chemical processes at the skin/patch boundary affect the delivery rate and may eventually inhibit drug delivery altogether.
The original performance target for controlled drug delivery is to achieve a zero-order release rate of the drug, so that a constant efficacious drug concentration is maintained in the blood plasma. However, more than two decades of research in chronobiology and chronopharmacology have demonstrated the importance of biological rhythms to the dosing of medications as well as determine the influence of a patient's circadian or other biological rhythms on drug efficacy and efficiency. This research reveals that certain disease symptoms follow a daily pattern, with peak symptoms at certain times of the day. It has been widely acknowledged that hormones, neurotransmitters and other intra-body compounds are released in different amounts at different times of the day pursuant to daily patterns.
The new approach stems from a growing body of research that demonstrates that certain diseases tend to get worse at certain times of the day. By synchronizing medications with a patient's body clock, many physicians believe that the drugs will work more effectively and with fewer side effects. In some cases, the improvements have been so pronounced that doctors have been able to reduce dosages. Circadian physiologic processes have been found to alter drug absorption, distribution, metabolism, and excretion. As a result, drag doses need to be adjusted to meet the differing needs of target organs or tissues at various times of the day (see, L. Lamberg, American Pharmacy, N831 (11): 20-23 (1991)).
The continued interest in chronopharmacology shows the ever-increasing need to develop technologies to control the temporal profile in drug delivery. Research findings suggest that the onset and severity of many diseases are cyclic in nature, or follow circadian patterns. Drag tolerance adds to the need for modulation of drag dosing profiles. Additionally, skin irritation and sensitization caused by medications may require intervals during which no drag is administered. Therefore, this improved form of drug delivery will be very important to people who need medicine easily, painlessly and automatically delivered to their bodies in timed increments (see Smolensk, M. H. & Lamberg, L. Body Clock Guide to Better Health: How to Use Your Body's Natural Clock to Fight Illness and Achieve Maximum Health, Henry Holt & Company, New York (2001) and Grimes, J.et al., J Pharmacol Exp Ther 285 (2): 457-463 (1998)).
Active transdermal delivery systems have been developed to help regulate the delivery rate by providing mechanisms to improve drag delivery over time by "pumping" the active ingredient. One such system, (U.S. Patent 5,370,635), describes a system for delivering a medicament and dispensing it to an organism for a relatively long period of time, for example at least a few days. The device can be adapted for positioning on the surface of the skin of a human or possibly an animal body in order to apply a medicament thereto from the outer side thereof. Conventional transdermal systems circumvent the disadvantages of the adsorption through the gastrointestinal tract, but they do not optimize or tailor the dosing regiment to offset peak symptoms. In addition the constant transdermal delivery of a drag can lead to severe side effects, including debilitating sleep disorders and ever increasing tolerance. A simple type of transdermal chronotherapy is a biphasic profile, in which the drug concentration changes from a high to a low level (or vice versa) over time. Although the system can be physically applied or removed to alter the drug level, patient compliance with this procedure may be difficult, particularly during inconvenient hours. To generate a biphasic profile, the delivery system may utilize an external regulator, as described in Fallon et al. (U.S. Patent 5,352,456, 1994) which illustrates a device for drug administration through intact skin that provides an initial pulse in the flux of the drug through the skin followed by a substantially lower flux of drug through the skin. Additionally, Fallon et al. (U.S Patent 5,820,875, 1998) later describe a device for the administration of a drug through an area of intact skin over a period of time in which the flux of the drug through the skin varies temporally in a controlled manner. The device is such that the skin flux of the drug varies in a controlled manner over the period of administration, typically from a high flux in the initial stage of administration to a lower flux in the later stage of administration.
Transdermal temporally controlled drug delivery systems, proposed by Giannos et al. (U.S. Patent 6,068,853, 2000) coupled pH oscillators with membrane diffusion in order to generate a periodic release of a drug or active ingredient transdermally, without external power sources and/or electronic controllers. The intent was to address chronotherapy with a pulsatile transdermal system. The strategy was based on the observation that a drug may be rendered charged or uncharged relative to its pKa value. Since only the uncharged form of a
drug can permeate across lipophilic membranes, including the skin, a periodic delivery profile may be obtained by oscillating the pH of the drug solution (see Giannos, S. A., "Pulsatile Delivery of Drugs and Topical Actives," in "Novel Topical Actives and Delivery Systems: Cosmetics, Dermatologicals and Transdermals", Edited by John. J. Wille, Jr.: Blackwell Publishing, Oxford UK (In press)). Recently, an orally administered drug for arthritis treatment has suggested a chronotherapeutic approach using a delay release system. The delay is scheduled to release the active ingredient at the beginning of an interleukin 6 cascade that is believed to cause early morning stiffness in rheumatoid arthritis patients. By attempting to synchronize the drug delivery with a biological cycle it is believed that low doses may be used to achieve desired results. However, this system does not overcome the limitations of delayed release systems described above.
Although it is possible to meet the requirements of chronopharmacology with pills, this requires an enormous amount of discipline by the patient to comply with the treatment regiment, see for example, U.S. Patent 6,214,379, which is incorporated herein by reference. As illustrated earlier, to achieve optimal results, many patients may need to wake up during the night to take their medication. Hence, what is needed is a non-invasive, reliable means of delivering drugs compounds in precisely timed and measured doses-without the inconvenience and hazard of injection, yet with improved performance as compared to orally delivered drugs.
Addressing patient compliance (taking the proper dosages at the prescribed times) is another critical problem facing caregivers and pharmaceutical firms alike. Studies show that only about half of patients take medications at the times and in the dosages directed by their physician. It is reported that each year, 125,000 deaths and up to 20% of all hospital and nursing home admissions result from patient noncompliance. It is estimated that non- compliance results in additional healthcare costs in excess of $100 billion per year in United States. These figures are even more pronounced for the elderly.
An individual's failure to comply with a dosing regimen, e.g. failure to take one or more doses of a drug or taking too many doses, will have an adverse impact upon the success of the regimen. Individuals may fail to comply with their drug dosing regimen for a number of reasons. For example, drug dosing regimens, such as every 4 hours, i. e. 8-12-4-8 involve a rigid dosing schedule that may be incompatible with an individual's personal schedule. Such a rigid dosing schedule when combined with normal human traits such as forgetfulness or denial of a medical condition, as well as a busy life, represent substantial obstacles to compliance with a drug dosing regimen. Accordingly, such rigid dosing regimens often result in the failure by an individual to take one or more doses at the prescribed time. This has an adverse impact on the levels of the therapeutic substance at the active site and consequently on the overall efficacy of the therapeutic substance. Hence, a need exists for systems and methods that increase patient compliance for administration of a variety of drugs.
Additional advantages and novel features of this invention shall be set forth in part in the description that follows, and in part will become apparent to those skilled in the art upon examination of the following specification or may be learned by the practice of the invention. The advantages of the invention may be realized and attained by means of the instrumentalities, combinations, compositions, and methods particularly pointed out in the appended claims.
Summary of the Invention
The present invention describes methods for treating diseases, addictions and disorders in humans. These methods involve synchronizing and tailoring the administration of drug compounds with the body's natural circadian rhythms, in order to counteract symptoms when they are likely to be at their worst, and are accomplished by using an automated and pre programmable transdermal drug administration system. This system can also utilize a pump or pressurized reservoir, and/or a system for removing depleted carrier solution, or other modulated dispensing actuator, in conjunction with micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers.
More specifically, these methods synchronize and tailor drug administration to the human body's circadian rhythms to deliver varying dosages at varying times. This ensures that peak drug concentrations are present in the bloodstream to offset peak disease and addiction symptoms arising from variances and fluctuation in the body's natural circadian rhythms. Further, these methods ensure that less of a drug is in the bloodstream when disease and addiction symptoms are at there lowest. This minimizes negative side effects, and increases efficacy of the dosing regimen. Brief Description of the Drawings
Figure 1 shows an exemplary device useful for implementing the present invention.
Figure 2A - 2B illustrate comparative drug release profiles demonstrating operation of the present invention.
Figure 3 is a schematic illustration of a drug delivery device in accordance with the present invention. Alternatively, permeation through the skin may be assisted by using micro- fabricated structures commonly referred to as Micro-needles, heating devices, iontophoretic devices, or sonophoretic devices that are attached to this device.
Figure 4 is a schematic illustration of an alternative drug delivery device in accordance with the present invention. Alternatively, permeation through the skin may be assisted by using micro-fabricated structures commonly referred to as Micro-needles, heating devices, iontophoretic devices, or sonophoretic devices that are attached to this device.
Figure 5 shows an exemplary administration profile for a stimulant delivery system.
Figure 6 shows an exemplary administration profile for a nicotine delivery system.
Figure 7 shows an exemplary administration profile for a nitroglycerin delivery system tailored to treat variant angina attacks. Figure 8 illustrates an exemplary administration profile for a nitroglycerin delivery system tailored to treat stress-induced angina attack.
Figure 9. illustrates an exemplary administration profile for an indomethacin delivery system tailored to arthritis.
Figure 10 illustrates an exemplary administration profile for a valdecoxib delivery system tailored to treat arthritis.
Figure 11 illustrates an exemplary administration profile for a tulobuterol delivery system tailored to treat asthma.
Figure 12 illustrates an exemplary administration profile for a clonidine delivery system tailored to treat hypertension.
Figure 13 illustrates an exemplary administration profile for a selegiline delivery system tailored to treat CNS degenerative disorders (Parkinson's Disease).
Figure 14 illustrates an exemplary administration profile for a selegiline delivery system tailored to treat Alzheimer's Disease and attention deficit disorder.
Figure 15 illustrates an exemplary administration profile for a methylphenidate delivery system tailored to treat ADD.
Figure 16 illustrates an exemplary administration profile for a selegiline delivery system tailored to treat depression.
Figure 17 illustrates an exemplary administration profile for an oxybutynin delivery system tailored to urinary incontinence.
Figure 18 illustrates an exemplary administration profile for a zolmitriptan delivery system tailored to treat migraine.
Figure 19 illustrates an exemplary administration profile for a miglitol delivery system tailored to treat diabetes. Figure 20 illustrates an exemplary administration profile for a fentanyl delivery system tailored to treat pain.
Figure 2 IA-C illustrates an exemplary administration profile for 5-fluorouracil, doxorubicin and cisplatin delivery system tailored to treat cancer.
Figure 22 illustrates an exemplary administration profile for a zidovudine delivery system tailored to treat AIDS.
Figure 23 illustrates an exemplary administration profile for a gabapentin delivery system tailored to epilepsy.
Figure 24 illustrates an exemplary administration profile for a triprolidine delivery system tailored to treat colds and flu.
Detailed Description of the Preferred Embodiments
Biological rhythms are periodic fluctuations in biological characteristics over time, which also include circadian as well as seasonal variations. The reality of circadian rhythms in animals including humans is well known (Halberg et al. J. Exp. Ther. Oncol. 3 (5) 223-260 (2003), Redfern et al. Chronobiology International 11 (4) 253-265 (1994)).
Circadian (approximately 24-hour) rhythms include the production of biological molecules such as endorphins, gonadotropin releasing hormone (GnRH), Cortisol and adrenaline. These regulate the body's temperature and heart rate, changes in characteristics of blood, such as stickiness, and behaviors such as wakefulness, sleep and periods of activity. Some of the rhythms that affect our bodies include:
• ultradian, which are cycles shorter than a day (for example, the milliseconds it takes for a neuron to fire, or a 90-minute sleep cycle)
• circadian, which last about 24 hours (such as sleeping and waking patterns)
• infradian, referring to cycles longer than 24 hours (for example monthly menstruation) • seasonal, such as seasonal affective disorder (SAD), which causes depression in susceptible people during the short days of winter.
Research demonstrates that certain disease symptoms follow a daily pattern, with peak symptoms at certain times of the day. It has been widely acknowledged that hormones, neurotransmitters and other intra-body compounds are released in different amounts at different times of the day pursuant to daily patterns. It is believed that the failure of current transdermal systems to synchronize drug administration with the body's natural rhythms often lead to (i) severe side effects, including debilitating sleep disorders (in the context of night¬ time nicotine administration, for example), (ii) ever increasing tolerance (in the case of nitroglycerin and other pharmaceuticals for example), (iii) more expensive therapies, due to the fact that more of a compound is needed because the daily body rhythm is ignored and time based dosing is not implemented.
In addition, many addictions follow a daily pattern consistent with one's circadian rhythms. For example, according to studies performed, immediately upon waking, smokers have peak nicotine cravings. These peak cravings return after each meal, due to the interplay of serotonin release as a trained response to the culmination of a meal. Our methods precisely time the administration of drugs so that they reach peak levels when symptoms are likely to be at their worst, and efficacy is greatly improved.
The present invention involves precisely timing the administration of drugs so that they reach peak levels in synchronization with times when symptoms are likely to be at their worst, or times at which the drugs are believed to be more effective in the body and/or better tolerated by the patient. The present invention is described in terms of a particular example of a drug delivery system that provides automated and precise control over dosing, with single-dose capability, (once while people sleep) or capability to administer separate and varying-sized doses many times throughout a multiple day period. The present invention also relates to the administration of different, distinct, drugs and dosages at different times of the day. The particular implementation is consistent with a commercial development of a miniaturized, automated and programmable non-invasive drug delivery system called the ChronoDose™ system being developed by the assignee of the present invention. The system enables controlling of the amount of drug exposed to the skin in a controlled time dependent way according to a programmed administration schedule that implements a desired dosage profile. In this manner the present invention enables one to precisely control and vary the time of drug release and the amount of each dose, pursuant to an easily set pre-programmed dosage profile. Research demonstrates that for certain symptoms, conditions and diseases, drug effects can be optimized when administered in a defined (and often varying) dosage at predefined times. This is known as Chronopharmacology (Reinberg, A. E., Concepts of Orcadian Chronopharmacology, In "Temporal Control of Drug Delivery" edited by Hrushesky, W.J.M., Langer, R. S. and Theeuwes, F. Annal NY Academy of Science, New York. Volume 618 102-115 (1991), Lemmer, B. Pharmacol Res. 33(2) 107-15 (1996)). To illustrate the importance of Chronopharmacology consider the following facts:
• Asthma attacks are 100 times more likely between 4:00 and 6:00 AM.
• Heart attacks and strokes are most likely to occur around 6:00 AM.
• Variant Angina attacks occur 30 times more often in the middle of the night between 2:00 AM and 4:00 AM.
• Smokers experience the highest cravings immediately upon waking up.
• Lethargy and difficulty getting out of bed is highest immediately upon waking up early in the morning.
• Cold and flu symptoms peak during nighttime and early morning hours, when cold medications are wearing off. In accordance with the present invention, substances with proven or suspected chrono-pharmacological efficiency are integrated into a miniaturized, automated, programmable watch-like device, such as device (100) shown in Fig. 1. The delivery system
(100) shown in Fig. 1 can be used for a variety of active compositions, and is small, fully automated and programmable. This system consists of a reusable wristwatch-like device
(101) to control the time and dosage of drug delivery; and a small, disposable, 'reservoir' (103), which is about the size of a quarter or 1/2 dollar coin in a particular example, or is cylindrical in shape, that the user can simply pop-in to place on the watch-like platform. This reservoir lasts, for example, up to 72 hours, depending on the application. Shorter and longer reservoir lifetimes are contemplated. The device is readily adapted to be worn on the forearm, ankle, or other convenient body location.
In a particular application the replaceable reservoir can include a description of an administration schedule that can be used to manually or automatically program device (100) with an administration schedule. For example, a written schedule can be printed on or affixed to the reservoir (101) or electrically programmed using volatile or non- volatile memory. In this manner, a dosing profile can be prescribed and filled by a pharmacy in much the same manner as a conventional drug prescription is handled today.
An exemplary implementation (shown in Fig. 3) comprises a collapsible drug reservoir, an expandable waste reservoir, a micro-pump, electronics for automation, a display, and a highly permeable membrane. Further, a heating element or a gas or air blowing apparatus may be used to assist evaporation of liquids into the waste reservoir or the environment. An exemplary system is described in a United Kingdom patent entitled Transdermal Drug Delivery and Method filed on September 13, 2004, Application Number PCT/IB2004/ 002947, which is incorporated herein by reference. The drug reservoir will contain between about 0.4 ml and 4 ml of drug formulation. A tiny, miniaturized pump is activated at pre-programmed times and releases a predefined amount of drug formulation into the drug chamber, where the formulation comes into contact with diffusion matrix. This diffusional matrix is in intimate contact with a highly permeable membrane. This membrane rests on the skin, and provides for even diffusion of the drug over the device's drug absorption surface area. This membrane works effectively with, and can be coated with, an adhesive, hydrogel or polymer substance, which allows for rapid transport kinetics. In operation, when the administration of the drug needs to be discontinued, the remaining drug formulation is either removed or evaporated from the membrane area via a waste chamber containing a desiccant, containing a hydrophilic substance (hydrogel) or the device is taken off. Further, to achieve chronopharmacological drug delivery for drugs that may not passively pass through the skin adequately, the above described device may use permeation enhancers whereby permeation through the skin is assisted, such as mechanical permeation enhancers that include micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers.
In an implementation shown in Fig. 4, a pressurized drug reservoir is used which minimizes or eliminates need for a micropump. Electronics control a valve that allows controlled quantities of the drug to be applied to the drug chamber where the formulation comes into contact with highly permeable membrane. Further, to achieve chronopharmacological drug delivery for drugs that may not passively pass through the skin adequately, the above described device may use permeation enhancers whereby permeation through the skin is assisted, such as mechanical permeation enhancers that include micro- fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, (together referred to as the Mechanical Permeation Enhancers) or a wide range of chemical permeation enhancers. The construction and use of transdermal patches for the delivery of pharmaceutical agents is known. See, for example, U.S. Patent 5,370,635, the disclosure of which is incorporated herein by reference. Such patches may be constructed using a saturated media, pressurized reservoirs, or unpressurized reservoirs with micropumps for continuous, pulsatile, or on-demand delivery of an active material.
For example, when administering a an active compound pursuant to a chronopharmacological dosage profile as set forth herein, using a programmed, transdermal, pulsatile drug delivery device, a pharmaceutically acceptable composition of an active material may be combined with either mechanical skin penetration enhancers including, but not limited to, micro-fabricated structures commonly referred to as Micro-needles, heat, iontophoresis, or sonophoresis, or a wide range of chemical permeation enhancers such as oleic acid, ethanol, amino acids, oleyl alcohol, long chain fatty acids, propylene glycol, polyethylene glycol, isopropanol, ethoxydiglycol, sodium xylene sulfonate, ethanol, N- methylpyrrolidone, laurocapram, alkanecarboxylic acids, dimethylsulfoxide, polar lipids, N- methyl-2-pyrrolidone, and the like, which increase the permeability of the skin to the active material and permit the active material to penetrate through the skin and into the bloodstream.
Pharmaceutically acceptable compositions may be combined with one or more agents including, but not limited to, alcohols, moisturizers, humectants, oils, emulsifiers, thickeners, thinners, surface-active agents, fragrances, preservatives, antioxidants, vitamins, or minerals.
Device-skin interface coupling media and/or control membranes include, but are not limited to, ethylcellulose, hydroxypropyl cellulose, poly (ethylene co-vinyl acetate), polyvinyl pyrrolidone, poly (ethylene oxide), poly (ethylene vinyl alcohol) and the like, to provide the composition in gel or hydrogel form, which may be dissolved in solvents such as water, methylene chloride or ethanol evaporated to the desired viscosity, and then applied to backing material to provide a patch. The control membranes can be any of the conventional materials such as microporous polyethylene, polyethylene co-vinyl acetate (EVA copolymer), polyurethane and the like.
Chronopharmacokinetics is defined as the predictable changes observed in the plasma levels of drugs and in the parameters used to characterize the pharmacokinetics of a drug. Studies on animals and humans indicate that the Cmax, Tmaχ, AUC and half-life often vary as a function of the hour of administration of the drug. Table 1 presents a list of medications for which temporal changes in pharmacokinetics have been documented.
Table 1. Drugs with documented time-dependent changes in pharmacokinetics*
Figure imgf000019_0001
* Labrecque, G. et al. Chronopharmacokinetics. Pharmaceutical News, 4 (2) 17-21 (1997)
We have carefully identified specific drugs and diseases because they have the following attributes: (i) Chronopharmacology is critical to optimized dosing but is not being implemented because no automated transdermal system exists, and (ii) these drugs can be transdermally absorbed passively (i.e., without the need for external modulation or pre- treatment such as sonophoresis, iontophoresis, electroporesis, microneedles, etc. or other permeation enhancement. Example substances include caffeine and ephedrine, and a variety of over-the-counter (OTC) and prescription stimulants (for treating fatigue, sleep disorders, attention deficit disorders and a variety of other conditions) in addition to herbal supplements, nicotine (for smoking cessation), nitroglycerin (for treating heart attack and strokes), fentanyl (for treating chronic pain), albutamol (for treating asthma), and selegiline (for treating depression, attention deficit disorder or Parkinson's disease). Exemplary chronopharmacological systems that can make use of the present invention are summarized in Table 2.
Table 2. Examples of disease states for ChronoDose™ application
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Using this system the present invention can preprogram the times and amount of each dosage by precisely controlling the amount of drug exposed to the skin during each dosing. This feature is advantageous when a drug is best administered during sleep, e.g., 1 to 2 hours before waking up. The present invention precisely counteracts peak disease symptoms and increase patient compliance.
The present invention represents the first true non-invasive chronopharmacological drug delivery device. While current transdermal applications are restricted to the dosage profile shown in Figure 2a, the automated implementation of the present invention can be programmed for a variety of drug delivery patterns to achieve customized patient dosing regiments for optimal therapy (Figure 2b).
There are many advantages for a controlled transdermal release of an active material such as a drug. As used herein, the term 'controlled' or 'sustained' release of an active material includes continuous or discontinuous, linear or non-linear release of the active material according to a programmed schedule. Among the advantages of controlled release are the convenience of a single application for the patient, avoidance of peaks and valleys in systemic concentration which can be associated with repeated injections, the potential to reduce the overall dosage of the active material, lower body stress, and the potential to enhance the pharmacological effects of the active material. A lower, sustained dose can also prevent adverse affects that are occasionally observed with infusion therapy. In addition to significantly reducing the cost of care, controlled release drug therapy can free the patient from repeated treatment or hospitalization, thus offering the patient greater flexibility and improving patient compliance.
A controlled release formulation of certain drugs also provides an opportunity to use the drug in a manner not previously exploited or considered. The present invention is particularly advantageous when (i) known chronopharmacological information indicates that a drug's effects can be optimized when administered in a defined dosage at a predefined time or times, and/or (ii) patient compliance with the dosing regimen is greatly increased due to automation, i.e. doses are required at inopportune times, i.e. at night while sleeping.
The present invention may be used to treat, cure, prevent, control or alleviate a wide range of conditions and symptoms. For example, the drug delivery regimen of the present invention is administered to treat a condition selected from the group consisting of vitamin and/or mineral deficiency, Cancer, Addiction, Arthritis, Parkinson's Disease, Attention Deficit Disorder, Cardiovascular Disorder, Cold/Flu Symptoms, Pain, Childhood Bronchial Asthma, Peptic Ulcer, Post-operative Recuperation, and so forth as shown below.
APPLICATIONS-ArisePatch™
A contemplated consumer product is the ArisePatch™. Most people experience difficulty and discomfort when waking early in the morning. According to a 2002 National Sleep Foundation poll 49 % of US adults age 18-29 have trouble waking in the morning and 41 % of US adults age 30-64 have trouble waking in the morning. There are 165,000,000 adults in the US alone age 18-64; meaning approximately 74,250,000 US adults age 18-64 have trouble waking in the morning. Chronotherapeutic Rationale:
The ArisePatch implementation of the present invention allows individuals, while asleep, to have an over-the-counter (OTC) or prescription stimulant automatically ad¬ ministered during a 1 -2 hour pre-wake-up period. Fig. 5 illustrates an exemplary stimulant administration profile showing a blood plasma level of ephedrine in nanograms per milliliter on the vertical axis, with time on the horizontal axis. Stimulant concentrations will reach peak levels immediately prior to having to wake. Immediately upon waking up the individual will be alert and feel well rested. The ArisePatch™ will eliminate the typical discomfort or difficulty associated with getting up early. This functionality is attractive to employed people getting up for work to ensure punctuality, and just about anyone who wants to offset morning discomfort associated with a late night, jet lag, or sickness.
APPLICA TIONS-Smoking Cessation (Example: Nicotine)
Nicotine replacement has been the most frequently used therapy to support smokers in their effort to quit. Smokers report that the craving for a cigarette is greatest immediately upon waking in the morning. The time elapsed between wakening and the first cigarette is the best indicator of addiction. For most smokers this time only a few minutes. Additionally, research has shown that nicotine transdermal delivery is influenced by chronopharmacokinetics. Nicotine patch design should compensate by decreasing the dose at night as well as increasing the dose in the morning and after meals (Gries et al., 1998).
Chronotherapeutic Rationale:
Current nicotine patches cause severe sleep disturbances by releasing nicotine steadily throughout the night to ensure sufficient morning nicotine levels to offset the strong morning craving. It is widely accepted that current nicotine patches have a detrimental and common side effect - sleeping disorders, and insomnia, including persistent nightmares. Therefore, users are often forced to remove the patch in the evening before they go to bed. This eliminates sleep disturbances, but results in nicotine levels that are insufficient to offset the strong morning craving. This is a major drawback to current nicotine patches and many users relapse, resulting in a less efficient smoking cessation therapy. Current patches present the user with a difficult decision, choosing between nightmares and relief from the strong morning cravings. Example:
An exemplary product contemplated by the present invention is called Nicotine ChronoDose™ system. In accordance with the present invention, the system can begin to administer nicotine (or nicotine analogs or any other smoking cessation compound including but not limited to bupropion) automatically during a one-hour period immediately prior to waking. This will relieve the smoker's peak craving upon waking without causing nightmares and insomnia. We believe that this system clearly provides a superior method for smoking cessation.
A more advanced nicotine replacement system than that described above is worn for three days at a time and is programmed to release nicotine in a daily rhythmic pattern such as shown in Fig. 6 to offset peaks in a smoker's cravings. Fig. 6 illustrates an exemplary nicotine administration profile showing a blood plasma level of nicotine in nanograms per milliliter on the vertical axis, with time on the horizontal axis. This implementation will reduce nicotine dependency by administering pre-programmed levels of nicotine pursuant to typical smoking patterns. For instance many smokers report that cravings for a cigarette are greatest upon waking up, after lunch, mid afternoon, after dinner and before bedtime. This implementation of the present invention will automatically release larger doses of nicotine to offset peak cravings and no nicotine when cravings are typically at a minimum. The present invention may be delivered in a preprogrammed manner for each treatment regimen. The only involvement by the user will be the replacement of the 'reservoir' every three days, and the replacement of the platform housing as needed.
This implementation represents a tremendous move forward in nicotine replacement therapy, and is far superior to the old-technology systems that simply release the same amount of nicotine all day and night. With the present invention, one can systematically decrease a smoker's tolerance without increasing dependence (the result of a constant flow) and better wean a smoker off nicotine. This will allow the smoker to better 'tailor-down' and decrease the amount of nicotine he needs to quit. Modern smoking cessation is much more than nicotine replacement therapy. Programs also include weight control, diet and psychological support. The present invention fits well into these programs, since it addresses the key component of being able to quit smoking by efficiently countering the withdrawal symptoms while doing away with the negative side effects of current nicotine replacement therapy systems, namely sleep disturbance.
APPLICA TIONS-Angina (Example: Nitroglycerin) Research shows that variant angina occurs 30 times more often between 2:00 a.m. and 4:00 a.m. ('critical angina phase') than at any other time of the day. Nitroglycerin effectively combats angina attacks, if administered in optimal doses. Current nitroglycerin patches exist, but they can only release a constant amount of nitroglycerin steadily over time. Current patches cannot tailor the release of nitroglycerin to optimize treatment by releasing more nitroglycerine precisely during the critical angina phase to offset these peak symptoms.
In addition, nitroglycerine loses its effectiveness and requires higher and higher dosages when administered constantly. Our bodies become tolerant to it. Current systems cannot stop or decrease the release of nitroglycerine when disease symptoms are lowest. Thus, these current 'dumb' patches cannot offset the critical angina phase by releasing more of the drug, nor can they shut down or stop nitroglycerin administration when the body doesn't need it. It is a 'one dose fits all' type of scenario once each "dumb" patch is applied to the patient. Chronotherapeutic Rationale:
The method in accordance with the present invention utilizes an automated transdermal system in order to transdermally administer more nitroglycerin during the critical angina phase to ensure adequate offset of these symptoms and less nitroglycerin when it is not needed so that no tolerance builds up. Our method utilizes a 'smart' patch medicine system at this time to offset these peak critical phases in the disease cycle arising due to the human body's circadian rhythm.
The pre-programmable automated transdermal system is worn around the wrist - like a watch (or the forearm arm or ankle) and releases nitroglycerin in optimal dosages at times that are optimally synchronized. This is pursuant to a pre-programmed and tailored dosage profile. Current nitroglycerin patches only have the capability to release a constant dose of nitroglycerin over a period of time. Current nitroglycerin patches simply cannot alter or vary dosages to increase dosages at different times of the day, and decrease dosages at other times of the day.
Example:
The nitroglycerin system in accordance with the present invention has three primary advantages over current nitroglycerin patches. First, the system automatically and precisely releases nitroglycerin in peak amounts to offset the peak symptoms of morning attacks occurring during the critical angina phase. Current nitroglycerin patches have release rates that stay constant and do not increase to offset critical phases, and do not decrease as symptoms decrease. Second, our system solves the tolerance issue by releasing less (or no) nitroglycerin in off-peak hours, and then releasing nitroglycerin at just the right time so that it is present during critical periods, without increasing tolerance. Third, our system accomplishes 1 and 2 above automatically, without the need for a patient to wake up to take a drug at this critical phase, which does away with the need for any increased patient compliance.
The nitroglycerin system represents an ideal delivery system for patients who use nitroglycerin regularly for the treatment and/or the prevention of heart attacks and strokes. Patient compliance regarding the timing and dose of heart attack medication is crucial. Patient non-compliance with physician's instructions for this is often a cause of re- hospitalization, according to the US Department of Health and Human Services. The system solves this problem, and will decrease the need for re-hospitalization by dramatically increasing patient compliance.
This system can be either an 'wear each night and remove in the morning' system, whereby it only releases nitroglycerin automatically to offset the critical angina phase in the morning, or a 'total solution' system, that is worn for a period of 24 hours to several days, and that administers nitroglycerin in tailored amounts and at tailored times as synchronized with the body's circadian rhythm (and conveniently taken off while showering or swimming).
The system is an innovative new drug therapy for angina. With the advantage of optimized and automated time and dose administration synchronized with a person's circadian rhythms, the system in accordance with the present invention ensures that nitroglycerin will circulate in the bloodstream exactly when the patient needs it, and without any build up tolerance. For these reasons, the present invention is superior to current steady release nitroglycerin patches. Our system's increased advantages are extremely relevant for those patients with moderate to severe angina.
Fig. 7 shows an exemplary administration profile for a nitroglycerin delivery system tailored to treat variant angina attacks or angina pectoris. This type of angina attack has a peak frequency in many patients between the hours of 2:00 and 4:00 AM. This is a particularly difficult time to wake up to take a drug such as nitroglycerin. In accordance with the present invention an administration profile substantially like that shown in Fig. 7 is automatically administered. In Fig. 7 the vertical axis indicates blood plasma level in nanograms per milliliter, and the horizontal axis indicates time from 10:00 PM through the night to 8:00AM.
Fig. 8 illustrates an exemplary administration profile for a nitroglycerin delivery system tailored to treat stress-induced angina attack. In Fig. 8 the vertical axis indicates blood plasma level in nanograms per milliliter, and the horizontal axis indicates time from 12:00 AM through the day until about 4:00 PM. The administration profile shown in Fig. 8 provides a high blood plasma concentration throughout the waking hours of a day when stress is likely occur.
APPLICATIONS: Arthritis (Examples: Indomethacin, Valdecoxib) An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automatically in the morning, just before a person awakes and the symptoms of arthritis are the worst. Later, towards mid-day, the drug concentration is also increased. Then in the evening, the drug dose is increased prior to bedtime.
Chronotherapeutic Rationale:
The most common forms, osteoarthritis and rheumatoid arthritis , both show distinctive circadian patterns of pain. While many people feel stiff for an hour or so after first getting up in the morning, people with osteoarthritis typically hurt most and have the most difficulty moving in the afternoon and evening. Those with rheumatoid arthritis almost always feel much worst in the morning. By dosing at night, early morning and mid-day, the benefits of non-steroidal anti-inflammatory drugs (NSAIDs) and cyclocoygenase-2 inhibitors (COX-2) can be maximized and side effects reduced. Examples of medications for arthritis include:
• Indomethacin (Indocin®)
• Diclofinac (Voltarin® and Cataflam®)
• Flurbiprofen (ANSAID®)
• Celecoxib (Celebrex®)
• Valdecoxib (Bextra®)
• Acetomenophen (Tylenol®)
• Oxaceprol
Example 1. Indomethacin (NSAID)
The primary adverse side effect of Indomethacin is gastrointestinal upset and bleeding. Therefore a transdermal arthritis patch would be a beneficial dosage form as opposed to oral tablets and capsules. Additionally, studies using indomethacin showed better efficacy and patient complience when dosed at night than when dosed at 8:00 am. Theoretical unenhanced transdermal flux for indomethacin (Berner-Cooper predictive model) is 0.93 ug/cm2/hr.
Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
5:00am-9:00am: BPC should be in the highest therapeutic range of between 0.5-2.0 mcg/ml. Peak 2 (Medium)
12:00pm to 8:00 pm: BPC should be in the medium therapeutic range of between 0.25 - 1.5 mcg/ml.
Peak 3 (Highest)
8:00pm- 11 :00pm: BPC should be in the highest therapeutic range of between 0.5 to 2.0 mcg/ml.
The time/dose chart should appear as shown in Figure 9 Example 2. Valdecoxib (COX-2 inhibitor)
Like indomethacin, the primary adverse side effect of COX-2 inhibitors is gastrointestinal upset and bleeding. Therefore a transdermal arthritis patch would be a beneficial dosage form as opposed to oral tablets and capsules. Lower blood plasma concentrations of COX-2 inhibors delivered transdermally has been suggested as therapeutically equivalent to higher BPC obtained by oral dosing.
Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest) 5:00am-9:00am: BPC should be in the highest therapeutic range of between 50-175 ng/ml. Peak 2 (Medium)
12:00pm to 8:00 pm: BPC should be in the medium therapeutic range of between 21-125 ng/ml.
Peak 3 (Highest)
8:00pm- 11:00pm: BPC should be in the highest therapeutic range of between 50 to 175 ng/ml.
The time/dose chart should appear as shown in Figure 10
APPLICA TIONS-Asthma (Example: Tulobuterol)
The automated transdermal asthma system automatically administers a morning dose of albuterol, tulobuterol, salmeterol, beta 2 agonist or any other antiarrhythmic drug (an 'Asthma drug') to combat the peak symptom of morning asthma attacks known as the 'morning dip'. Chronotherapeutic Rationale:
Asthma attacks occur 100 (one hundred) times more often between the hours 4 A.M. and 6 A.M., when most people are asleep. This is due to the early morning deterioration of respiratory function known as 'morning dip', which is the time of day that respiratory function is at its lowest. These early morning asthma attacks cause great distress to sufferers and care providers. The morning dip represents the dip in respiratory function at this time when asthma attacks are 100 times more likely to occur. Our system effectively combats the morning dip by releasing more Asthma drug at this time to offset this peak morning symptom. In other words, our 'smart' patch varies the level of drug in the bloodstream so that drug concentrations are highest when respiratory function is at its lowest.
Current 'dumb' asthma patches exist, but they can only release a constant amount of drug steadily over time. Current patches cannot tailor the release of drug to optimize treatment by releasing more drug precisely during the morning dip to offset these peak critical symptoms.
The Asthma system has two primary advantages over current patches. First, the system of the present invention utilizes its core competitive advantage to automatically and precisely release tulobuterol or other asthma drugs in peak amounts to offset the peak symptoms associated with the morning dip. Current patches have release rates that stay constant and do not increase to offset this peak critical phases, and do not decrease as symptoms decrease. Second, our system accomplishes 1 and 2 above automatically, without the need for a patient to wake up to take a drug at this critical phase, which does away with the need for any increased patient compliance.
The automated transdermal system for asthma is worn around the wrist like a watch (or the forearm arm or ankle) and releases albuterol or other asthma drugs in optimal dosages at times that are optimally synchronized, especially to offset the morning dip, pursuant to a pre-programmed and tailored dosage profile. Current Asthma patches only have the capability to release a constant dose over a period of time. Current Asthma patches simply cannot alter or vary dosages to increase dosages at different times of the day, and decrease dosages at other times of the day.
The system is an innovative new drug therapy for asthma. With its superior advantage of optimized and automated time and dose administration synchronized with our circadian rhythms, our system ensures that tulobuterol or another asthma drug will circulate in increased amounts in the bloodstream exactly when the patient needs it. For these reasons, our system is superior to current steady release patches. Our system's increased advantages are extremely relevant for those patients with moderate to severe asthma. The time/dose chart should appear as shown in Figure 11
APPLICATIONS-Hypertension (Example: Clonidine)
Current clonidine patches release the drug consistently over time. It cannot release more of the drug when symptoms are worst. People die most when the symptoms peak. Having the advantage of administering more of the drug when a patient needs it the most can mean the difference between life and death, especially in patients with moderate to severe high blood pressure. Chronotherapeutic Rationale:
The automated transdermal system for hypertension has two primary advantages over current patches. First, our system utilizes its core competitive advantage to automatically and precisely release clonidine or other hypertension drugs in peak amounts to offset the peak symptoms associated with the dangerous morning symptoms. Current hypertension patches have release rates that stay constant and do not increase to offset this peak critical phases, and do not decrease as symptoms decrease. Second, our system accomplishes 1 and 2 above automatically, without the need for a patient to wake up to take a drug at this critical phase, which does away with the need for any increased patient compliance. The clonidine automated transdermal system utilizes clonidine, (or another hypertension drug) an effective drug that combats high blood pressure. Example:
The clonidine automated transdermal drug delivery system has an automated morning release of Clonidine to combat the peak symptom of morning heart attacks. Blood pressure differs at different times of the day. Blood pressure surges upon waking, and is lower by 20 to 30 per cent while sleeping. Our preprogrammed automatic transdermal system utilizes its core competitive advantage by releasing clonidine in a tailored fashion to counter high blood pressure when symptoms are highest, while releasing less clonidine when symptoms are less severe. The time/dose chart should appear as shown in Figure 12
APPLICATIONS-CNS Degenerative Disorders
(Example: Selegiline) Parkinson 's Disease
Sleep disturbances in Parkinson's disease patients reveal alterations of circadian rhythms. Autonomic dysfunction, described in Parkinson's disease, reveals numerous alterations in circadian regulations including loss of circadian rhythm of blood pressure, increased diurnal blood pressure variability, and postprandial hypotension. Many biologic indices such as Cortisol, catecholamines, and melatonin are also altered. Circadian rhythms in dopaminergic systems as well as possible daily fluctuations in kinetics of drug treatments are likely involved in such variations. Chronotherapeutic Rationale:
Primary negative side effects of the selegiline patches are abnormal dreams, insomnia, and difficulty sleeping. We believe that by specifically refraining from administering selegiline at night, and utilizing our system's core competitive advantage to turn it on an hour or so before waking, we can do away with this negative side effect and still offset the critical phase of morning symptoms of depression. It has been reported that patients have increased symptoms of depression upon waking if the critical amount of Selegiline is not circulating through their system.
The selegiline automated transdermal drug delivery system gives an automated morning release of selegiline to combat the peak symptom of morning depression without the side effect of sleep disturbances. The system in accordance with the present invention is applied before bed. It does not release the drug until one or two hours before morning, so symptom of morning depression would be corrected by our system without subjecting the patient to sleep disturbances. The time/dose chart should appear as shown in Figure 13
Alzheimer's disease
Selegiline is an effective MAO inhibitor for the treatment of depression, Alzheimer's and Attention Deficit Disorder. Currently oral selegiline produces many undesirable side effects. A transdermal form of selegiline, EMSAM™, is currently being developed. However, it also produces sleep disturbances as well. It is believed that the system in accordance with the present invention would be superior to conventional selegiline product delivery systems. Chronotherapeutic Rationale:
Primary negative side effects of the selegiline patches are abnormal dreams, insomnia, and difficulty sleeping. We believe that by specifically refraining from administering selegiline at night, and utilizing our system's core competitive advantage to turn it on an hour or so before waking, we can do away with this negative side effect and still offset the critical phase of morning symptoms of depression. It has been reported that patients have increased symptoms of depression upon waking if the critical amount of Selegiline is not circulating through their system.
The selegiline automated transdermal drug delivery system gives an automated morning release of selegiline to combat the peak symptom of morning depression without the side effect of sleep disturbances. The system in accordance with the present invention is applied before bed. It does not release the drug until one or two hours before morning, so symptom of morning depression would be corrected by our system without subjecting the patient to sleep disturbances.
The time/dose chart should appear as shown in Figure 14 APPLICATIONS-Attention Deficit Disorder (Example: Methylphenidate)
Ritalin is indicated as an integral part of a total treatment program which typically includes other remedial measures (psychological, educational, social) for a stabilizing effect in children with a behavioral syndrome characterized by the following group of developmentally inappropriate symptoms: moderate-to-severe distractibility, short attention span, hyperactivity, emotional lability, and impulsivity.
Methylphenidate is usually administered in divided doses 2 or 3 times daily, preferably 30 to 45 minutes before meals. Patients who are unable to sleep if medication is taken late in the day should take the last dose before 6 p.m. Since the suggested first dose is early in the morning, it would be beneficial to automatically control the dosage.
Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
6:00am-8:00am: BPC should be in the highest therapeutic range of between 8-25 ng/ml. Peak 2 (Highest)
10:00am to 12:00 pm: BPC should be in the highest therapeutic range of between 8-25 ng/ml.
Peak 3 (Highest)
3:00pm- 5:00pm: BPC should be in the highest therapeutic range of between 8 to 25 ng/ml. The time/dose chart should appear as shown in Figure 15
APPLICA TIONS-Depression (Example: Selegiline) Selegiline is an effective MAO inhibitor for the treatment of depression, Alzheimer's and Attention Deficit Disorder. Currently oral selegiline produces many undesirable side effects. A transdermal form of selegiline, EMSAM™, is currently being developed. However, it also produces sleep disturbances as well. It is believed that the system in accordance with the present invention would be superior to conventional selegiline product delivery systems. Chronotherapeutic Rationale:
Primary negative side effects of the selegiline patches are abnormal dreams, insomnia, and difficulty sleeping. We believe that by specifically refraining from administering selegiline at night, and utilizing our system's core competitive advantage to turn it on an hour or so before waking, we can do away with this negative side effect and still offset the critical phase of morning symptoms of depression. It has been reported that patients have increased symptoms of depression upon waking if the critical amount of Selegiline is not circulating through their system.
The selegiline automated transdermal drug delivery system gives an automated morning release of selegiline to combat the peak symptom of morning depression without the side effect of sleep disturbances. The system in accordance with the present invention is applied before bed. It does not release the drug until one or two hours before morning, so symptom of morning depression would be corrected by our system without subjecting the patient to sleep disturbances. The time/dose chart should appear as shown in Figure 16
APPLICATIONS: Urinary Incontinence (Example: Oxtybutynin)
An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automatically at night while asleep, and to decrease concentrations during the daytime work hours, and again to slightly increase drug concentrations after work and prior to bed.
Chronotherapeutic Rationale:
The primary adverse side effect of Oxybutynin is daytime sleepiness, daytime attention and cognitive deficits, drowsiness, dizzyness, blurred vision, (must use caution when driving, operating machinery, or performing other hazardous activities). Therefore, it seems that a dose in the lower end of the therapeutic range should be administered during the daytime, with a slightly larger dose administered after working hours, and with an even higher dose administered during the sleeping hours.
This would reduce the potentially serious adverse side effect of daytime drowsiness and daytime cognitive impairment. This dosing regimen would also give the user a higher dose at night, when one sleeps. At this time, increased drowsiness would be advantageous as well as providing a period of undisturbed sleep due to the inhibition of urge incontinence.
Medications for incontinence include:
• Oxybutynin (Ditropan® and Oxytrol®)
• Tolterodine (Detrol®)
• Duloxetine (Yentreve®)
Example 1. Oxybutynin
The mean maximum blood plasma concentration following oral dosing with 5 mg oxybutynin or transdermally with 39 mg is 3 ng/mL. Blood plasma concentration between 1 and 3 ng/ml
Theoretical unenhanced transdermal flux for oxybutynin (Berner-Cooper predictive model) is 10.98 ug/cm2/hr.
NOTE: Dose of current Oxytrol patches are 3.9 mg per day. Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
11 :00pm-7:00am: BPC should be in the highest therapeutic range of between 2.5-4.5 ng/ml. Peak 2 (Low)
7:00am to 5:00 pm: BPC should be in the lowest therapeutic range of between 0.75 - 1.5 ng/ml.
Peak 3 (Medium)
5:00pm- 11 :00pm: BPC should be in the medium therapeutic range of between 1.5 to 2.5 ng/ml. The time/dose chart should appear as shown in Figure 17
APPLICATIONS: Headache and Migraine (Example: Zolmitriptan)
An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automaticaly in the evening to provide needed medication, in the very early morning (0200 - 0400) while asleep, and again later on (0800 - 1000) upon waking. Then, during the daytime work hours, decrease concentrations to allow for normal activities. Chronotherapeutic Rationale:
Migraine, cluster and tension-type headaches may produce a headache that awakens an individual in the early morning hours (usually between 2 and 4 AM), or is present upon awakening. Those individuals with chronic tension-type headache are most likely to be awakened in the early morning hours due to headache. This headache also tends to be at its worst severity at that time of day. A variety of causes may account for this early-morning pattern to the headaches.
Additionally, primary headaches associated with late sleeping or weekends are caused by caffeine withdrawal. Sleeping in late delays morning caffeine intake, which often leads to withdrawal and migraine. Many people reduce their caffeine intake on weekends, which readily explains the weekend increase in migraine attacks. Fewer migraines occur on Mondays and Tuesdays than on other days of the week.
Medications for headache and migraine include: Abortive Medications
• Analgesics with caffeine such as Excedrin® Migraine (acetaminophen, aspirin and caffeine).
• Analgesics with caffeine and barbiturates such as Fiorinal® (butalbital, aspirin and caffeine) and Fioricet®(butalbital, acetaminophen and caffeine).
• Non steroidal antiinflammatory drugs (NSAIDs) such as Advil® (ibuprofen), and Aleve® (naproxen sodium).
• Ergotamines such as Cafergot® (caffeine and ergotamine tartrate) and Migranal® (dihydroergotamine).
• Triptans such as Zomig®(zolmitriptan), Maxalt® (rizatriptan), Imitrex® (sumatriptan), Frova® (frovatriptan), Axert® (almotriptan) and Amerge® (naratriptan).
Excedrin Migraine is a registered trademark of Bristol-Myers Squibb Company
Fiorinal and Fioricet are registered trademarks of Novartis Pharmaceuticals Corporation
Advil is a registered trademark of Whitehall-Robbins Healthcare
Aleve is a registered trademark of Bayer Corporation
Cafergot and Migranal are registered trademarks of Novartis Pharmaceuticals Corporation Zomig is a registered trademark of AstraZeneca
Maxalt is a registered trademark of Merck & Co., Inc.
Imitrex is a registered trademark of GlaxoSmithKline
Frova is a registered trademark of Elan Pharmaceuticals/UCB Pharma, Inc.
Axert is a registered trademark of Pharmacia
Amerge is a registered trademark of GlaxoSmithKline
Preventive Medications
• Beta blockers such as Inderal® (propranolol)*, Blocadren ® (timolol maleate)*, and metoprolol.
• Calcium-channel blockers such as Cardizem® (diltiazem) and Procardia® (nifedipine).
• Antidepressants such as Prozac® (fluoxetine), Paxil® (paroxetine) and Zoloft® (sertraline).
• Anticonvulsants such as Depakote® (valproic acid or divalproex sodium).*
• NSAIDs such as Orudis® (ketoprofen) and Aleve® (naproxen sodium). Inderal is a registered trademark of AstraZeneca
Blocadren is a registered trademark of Merck & Co, Inc.
Cardizem is a registered trademark of Aventis Pharmaceuticals
Procardia is a registered trademark of Pfizer Inc.
Prozac is a registered trademark of Eli Lilly and Company
Paxil is a registered trademark of GlaxoSmithKline
Zoloft is a registered trademark of Pfizer Inc.
Depakote is a registered trademark of Abbott Laboratories
Orudis is a registered trademark of Aventis Pharmaceuticals
Aleve is a registered trademark of Bayer Corporation Example, Zolmitriptan
Blood plasma concentration between 1.0 and 5.0 ng/ml
Theoretical unenhanced transdermal flux for zolmitriptan (Berner-Cooper predictive model) is 6.02 ug/cm2/hr.
Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times:
Peak 1 (Highest)
2:00am-4:00am: BPC should be in the highest therapeutic range of between 3.5-4.0 ng/ml.
Peak 2 (Highest)
8:00am- 10:00am: BPC should be in the highest therapeutic range of between 3.5-4.0 ng/ml.
Trough (Lowest)
12:00pm to 12:00 am: BPC should be in the lowest therapeutic range of between 1.0- 3.0 ng/ml.
The time/dose chart should appear as shown in Figure 18
APPLICATIONS: Diabetes (Example: Miglitol)
An automated, and programmed, pulsatile drug delivery regimen is desired to in order to increase drug concentrations automaticaly in the morning (0800), midday (1200) and evening (1800) which coincide with mealtimes.
Miglitol is indicated as an adjunct to diet to improve glycemic control in patients with non-insulin-dependent diabetes mellitus (NIDDM) whose hyperglycemia cannot be managed with diet alone. Theoretical unenhanced transdermal flux for miglitol (Berner-Cooper predictive model) is 49.24 ug/cm2/hr.
Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
8: 00am- 10:00am: BPC should be in the highest therapeutic range. Peak 2 (Highest)
12:00am- 2:00pm: BPC should be in the highest therapeutic range. Trough (Highest)
6:00pm to 8:00 am: BPC should be in the lowest therapeutic range.
The time/dose chart should appear as shown in Figure 19
APPLICA TIONS-Pain Management
(Example: Fentanyl
Many diseases and pain-causing situations (post-surgery, post trauma) have predictable pain patterns. For example, Cortisol is virtually absent in the body overnight, and this is what fights inflammation. Thus, any pain resulting from inflammation (rheumatoid arthritis, post-surgical pain, post-traumatic pain, back pain, neurological pain) is most common in the early morning hours between 3:00 a.m. and 8:00 a.m. Migraine pain is worst around 6:00 a.m. Ankylosing spondylitis pain surges between 6:00 a.m. and 9:00 a.m. Osteoarthritis pain surges in mid-afternoon.
Pain varies tremendously from one patient to the next, and there are also some studies suggesting that the intensity of pain varies according to time of day. In human studies, pain induced experimentally was reported to be maximal in the morning, or in the afternoon or at night. A circadian pattern of pain has been seen in patients suffering from pain produced by different diseases. For instance, highest toothache intensity occurred in the morning, while biliary colic, migraine, and intractable pain were highest at night. Patients with rheumatoid anhritis reported peak pain early in the morning, while those with osteoarthritis of the knee indicated that the maximal pain occurred at the end of the day. The effectiveness of opioids appears also to vary according to time of day, but large differences in the time of peak and low effects were found. Peak pain intensity and narcotic demands occur early in the morning, or it can be at the end of the day. Pain is a complex phenomenon and specific to each clinical situation.
An automated, and programmed, pulsatile transdermal drug delivery regimen is needed to substantially increase blood plasma concentrations of Fentanyl or other pain medications, automatically between 3:00am and 8:00 am, while people sleep, where pain results from inflammation, because cortisone, a key inflammation fighter, is lowest in the body at that time. Additionally, an automated, and programmed, pulsatile transdermal drug delivery regimen is needed to substantially increase blood plasma concentrations of Fentanyl or other pain medications automatically between 6:00am and 9:00am for Ankylosing spondylitis pain, and in mid-afternoon for Osteoarthritis pain.
Other pain medication includes: codeine, dihydrocodeine, hydrocodone or hydromorphone, Sufentanil, Nalbuphine, Buprenorphine, Hydromorphone and any type of opiate derivative.
These are exemplary choices for transdermal pain management since they are effective, there is considerable hepatic first pass effect and a short half life, and they are skin permeable.
For example, for pain that increases with inflammation, as in the situations noted above, our regimen would suggest automated and programmed, transdermal pulsatile delivery of fentanyl to reach blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
3:00am-8:00am: BPC of fentanyl should be in the highest therapeutic range of between 2-8ng/ml. Peak 2 (Lowest)
8: 00am- 5: :00pm: BPC should be in a moderate therapeutic range of between l-3ng/m. Peak 3 (Middle)
5:00pm to 3:00am: BPC should be in the lowest therapeutic range of between 2- 5g/ml.
The time/dose chart should appear as shown in Figure 20
APPLICA TIONS-Cancer Example:
Cancer chronotherapy is attracting attention as a novel and logical therapy in which anti-cancer drugs are administered with optimal timing according to circadian rhythms of anti-cancer action and those of adverse effects on normal cells. Advances in chronobiology have identified the suprachiasmatic nucleus (SCN) as the center of biological rhythms and the area in which clock genes such as PERl, PER2, PER3, CLOCK, BMALl, TIM, CRYl, CRY2, tau act to generate and coordinate biological rhythms. These findings have led to the development of chronotherapy. Clinically, patients with advanced gastrointestinal cancer have been treated by chrono-modulated chemotherapy with good response. For colorectal cancer patients with un resectable liver metastases, chronotherapy with g-OHP + 5-FU + FA (folinic acid) has been reported to allow complete surgical resection of liver metastases, resulting in 39—50% 5-year survival. The circadian timing of surgery, anticancer drugs, radiation therapy, and biologic agents can result in improved toxicity profiles, tumor control, and host survival. Optimally timed cancer chemotherapy with doxorubicin or pirarubicin ( 06:00 h) and cisplatin ( 18:00 h) enhanced the control of advanced ovarian cancer while minimizing side effects, and increased the response rate in metastatic endometrial cancer. Therapy of metastatic bladder cancer with doxorubicin-cisplatin was made more tolerable by this same circadian approach resulting in a 57% objective response rate. This optimally timed therapy is also effective in the adjuvant setting, decreasing the expected frequency of metastasis from locally advanced bladder cancer. Circadian fluorodeoxyuridine (FUDR) continuous infusion (70% of the daily dose given between 15:00 h and 21 :00 h) has been shown effective for metastatic renal cell carcinoma resulting in 29% objective response and stable disease of more than 1 yr duration in the majority of patients. Toxicity is reduced markedly when FUDR infusion is modulated to circadian rhythms
Chronotherapy has also been used to lower the amount of side effects from chemotherapy drugs. Over the years, doctors have realized that by giving two of these drugs, Adriamycin and cisplatin, in the morning and evening, respectively, side effects could be cut in half.
Thus, dosing could be optimized using the ChronoDose system. For example, pulsatile delivery should have blood plasma concentrations (BPC) as set forth for each specific medication.
The time/dose charts should appear as shown in Figure 21 (a, b & c)
APPLICATIONS- Acquired Immune Deficiency Syndrome (AIDS/HIV) Examples: Zidovudine, Didanosine
Currently available antiretroviral drug regimens are able to suppress HIV replication and allow CD4 recovery in the vast majority of patients with HIV infection. The challenge is to match each patient to the regimen that is most likely to durably suppress HIV replication enough to prevent resistance selection without causing treatment-limiting toxicities. It is also critical, but difficult, to know when to begin treatment relative to CD4 cell count and plasma viral load.
Adherence to antiretroviral therapy for the treatment of HIV infection and AIDS has become one of the most important clinical challenges among HIV health care providers and patients. Adherence to the prescribed regimen may predict which patients achieve undetectable viral loads. Unfortunately, non-adherence is common in antiretroviral therapy and has been associated with increases in viral load and the development of drug resistance. Efforts to maximize patient adherence are critical for suppressing HIV replication and preventing the transmission of drug-resistant virus.
Automated and programmed, transdermal pulsatile delivery of zidovudine to reach blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
5:00am-9:00am: BPC of zidovudine should be in the highest therapeutic range. Peak 2 (Highest)
7:00pm to 11 :00pm: BPC should be in the highestest therapeutic range.
Theoretical unenhanced transdermal flux for zidovudine (Berner-Cooper predictive model) is 17.94 ug/cm2/hr.
The time/dose chart should appear as shown in Figure 22
APPLICA TIONS-Epilepsy Example: Gabapentan
In the majority of persons with the brain disorder epilepsy, seizures recur at predictable times of day. About half of those with epilepsy experience seizures mainly in waking hours. About one-quarter have them mainly in sleep. In the others, timing is less consistent; their seizures strike both day and night.
More than twenty anti-seizure medications (also called anticonvulsant or anti-epilepsy drugs) currently are available in the United States. Some are specifically designed not to interfere with the activity of other drugs, including birth control pills. They include gabapentin (Neurontin), lamotrigme (Lamictal), topiramate (Topamax), tiagabine (Gabatril), levetiracetam (Keppra), and oxcarbazepine (Trileptal).
None of the newer medications and only two of the older ones, valproate and phenytoin, have been studied with regard to how they work when taken at different times of the day or in different phases of the menstrual cycle. Whether the findings in valproate and phenytoin can be generalized to other anti-epilepsy drugs is not known; the results do raise issues, however, that urgently need further study. Studies of valproate show that people absorb it more slowly and less efficiently when they take it in the evening than in the morning. This finding is of concern because protection against seizures usually is needed most in NREM sleep, the state that dominates the first half of a night's sleep.
Automated and programmed, transdermal pulsatile delivery of gabapentan to reach blood plasma concentrations (BPC) as set forth below within the following ranges at the following times: Peak 1 (Highest)
5:00am-9:00am: BPC of gabapentan should be in the highest therapeutic range. Peak 2 (Highest)
7:00pm to 11 :00pm: BPC should be in the highestest therapeutic range.
The time/dose chart should appear as shown in Figure 23
APPLICATIONS-CoId and Flu treatment Example: Triprolidine
Cold and flu symptoms are worst from midnight until the early morning because the concentration of Cortisol is lowest at that time. Current nighttime cold and flu medication end up losing efficacy by early morning when cold and flu symptoms are highest. Therefore people suffering from a cold or flu are often unpleasantly awoken by an increase in symptoms, cutting sleep short. Set and put on before bedtime, the present invention will automatically deliver a larger dose of medication and immuno-boosters in the early morning hours to more effectively combat the peak cold and flu symptoms that occur in the morning.
This implementation uses prescription or OTC cold medicine alone or optionally in combination with certain transdermally efficacious vitamins and immune system boosters to provide a total solution to cold and flu ailments. This is the first cold therapy that combines OTC medicine with supplemental immuno-boosters in a comprehensive and automated manner.
In a particular application, the Cold and Flu automated transdermal drug delivery system utilizes OTC cold medicine, Vitamin C, Echinacea, and Zinc to provide a total solution to cold and flu ailments, and all while a person sleeps. The Cold/Flu system releases these combination of compounds every 2 hours throughout the night, with a higher dosage of compounds being released in the morning to combat these proven middle of the night and early morning symptoms, which are the worst of the day. Users will experience less severe cold and flu symptoms during the morning hours, will not have their sleep cycle cut short, and will wake up feeling symptom-free.
The time/dose chart should appear as shown in Figure 24
APPLICATIONS-Weight Control, Vitamin and Herbal Supplementation In yet another application, a series of weight loss vitamins and supplements is administered in small distinct doses many times over several days. Vitamins and supplements are absorbed by the body in small dosages. Contrary to popular belief, once-a- day products are not maximally effective because excess dosages are excreted unused. This implementation of the present invention precisely controls the timing and dosage of small but distinct amounts of vitamins and supplements during a 24-hour period to ensure that vitamins and supplements are constantly bio-available for optimal absorption and cellular function. Greater doses are automatically released prior to mealtimes to counter appetite cravings, resulting in a much more effective diet program.
APPLICATIONS-In general
The present invention is particularly useful in applications in which it is necessary and/or desirable to start the administration of a drug, stop the administration of a drug, and/or increase/decrease the dosage of a drug at a time when it is inconvenient or impossible for a patient to initiate the necessary actions. This is particularly useful for a wide variety of drug administration applications that benefit when an administration is started, stopped, or changed while a person is sleeping. As research and knowledge of chronotherapy increases, it is contemplated that a wide variety of applications will be discovered in which benefit is realized by starting, stopping and/or changing the drug administration while a patient sleeps.
In each of the examples, treatment is continued as needed to provide superior symptomatic relief, prevent exacerbation of symptoms, and/or prevent and/or delay progression of the disease state or condition in the patient, or until it is no longer well tolerated by the patient, or until a physician terminates treatment. For example, a physician may monitor one or more symptoms and/or serum levels of active material and/or metabolic by-product(s) in a patient being treated according to this invention and, upon observing attenuation of one or more symptoms for a period of time, conclude that the patient can sustain the positive effects of the above-described treatment without further administration for a period of time. When necessary, the patient may then return at a later point in time for additional treatment as needed.
As used herein, 'day' means a 24-hour period. Thus, for example, 'for at least three consecutive days' means for at least a 72-hour period. During or after the treatment, a physician may monitor one or more symptoms and/or serum levels in the patient and, upon observing an improvement in one or more of the parameters for a period of time, conclude that the patient can sustain the positive effects of the treatment without further administration of the active material for a period of time.
In order to use an active material for therapeutic treatment (including prophylactic treatment) of mammals including humans according to the methods of this invention, the active material is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition. According to this aspect of the invention there is provided a pharmaceutical composition comprising an active material in association with a pharmaceutically acceptable diluting substance or carrier, wherein the active material is present in an amount for effective treating or preventing a particular condition.
While individual needs may vary, determination of optimal ranges for effective amounts of an active ingredient (alone or in combination with other drugs) within the ranges disclosed herein is within the expertise of those skilled in the art. Accordingly, 'effective amounts' of each component for purposes herein are determined by such considerations and are amounts that improve one or more active ingredient functions and/or ameliorate on or more deleterious conditions in patients and/or improve the quality of life in patients.
Pharmaceutical Kits
The present invention also provides pharmaceutical kits for treating a particular symptom, condition and/or disease and/or improving a particular biological function, comprising one or more containers comprising one or more active compositions in accordance with this invention. Such kits can also include additional drugs or therapeutics for co-use with the active composition for treatment or prevention of a particular symptom, condition and/or disease and/or improving a particular biological function. In this embodiment, the active composition and the drug can be formulated in admixture in one container, or can be contained in separate containers for simultaneous or separate administration. The kit can further comprise a device(s) for ad-ministering the compounds and/or compositions, such as device 100 shown in Fig. 1, and written instructions in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which instructions can also reflect approval by the agency of manufacture, use or sale for human administration. Although the invention has been described and illustrated with a certain degree of particularity, it is understood that the present disclosure has been made only by way of example, and that numerous changes in the dosages, administration profiles, timing, as well as the combination and arrangement of parts can be resorted to by those skilled in the art without departing from the spirit and scope of the invention, as hereinafter claimed.

Claims

We claim
1. A method for delivering a bioactive agent to a human or animal comprising: providing a trans dermal drag delivery device coupled to the human or animal, the delivery device having a source of the bioactive agent, a programmable timing mechanism, and a mechanism for causing the bioactive agent to be delivered transdermally in response to the timing mechanism; and timing routines implemented by the timing mechanism, wherein the timing routines are selected to deliver the bioactive agent at a time, rate, sequence and/ or cycle that is synchronized with a biological rhythm of the human or animal.
2. The method of claim 1 whereby permeation through the skin is assisted using micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, or a wide range of chemical permeation enhancers.
3. The method of claim 1 wherein the bioactive agent comprises a stimulant and the timing routines are selected to deliver the stimulant immediately before the human or animal wakes up.
4. The method of claim 1 wherein the bioactive agent comprises nicotine and the timing routines are selected to deliver the nicotine at times that are associated with nicotine cravings.
5. The method of claim 3 wherein at least one of the selected times corresponds to a time at which the human or animal experiences a morning nicotine craving.
6. The method of claim 1 wherein the bioactive agent comprises an antihistamine and the timing routines are selected to deliver the antihistamine while the human or animal sleeps.
7. The method of claim 1 wherein the bioactive agent comprises a stimulant and the timing routines are selected to deliver the stimulant immediately before the human or animal wakes up.
8. A method for treating a symptom, condition, and/or disease comprising: a) identifying a drag suitable for treating a particular symptom, condition and/or disease; b) identifying a biologically superior time for administering a drug; c) programming a time-programmable transdermal drug delivery system with a schedule selected to synchronize with the identified biologically superior time; and d) causing the time-programmable transdermal drug delivery system to deliver the active ingredient according to the programmed schedule.
9. A programmable transdermal drug delivery device comprising: a) an interface for coupling to the skin of a host; b) a reservoir storing a quantity of an active composition; c) valve mechanism for supplying a quantity of the active composition from the reservoir to the interface in response to a control signal; and d) a timing mechanism coupled to the valve mechanism and configured to generate the control signal according to a programmed administration schedule.
10. The device of claim 7 wherein the valve mechanism controls a rate at which the active composition is supplied in response to the control signal.
11. The device of claim 7 further comprising a mechanism for removing the active composition from the interface in response to the control signal.
12. The device of claim 7 further comprising a mechanism for removing carrier materials from the interface.
13. A kit comprising a first container comprising a pharmaceutical formulation of an active material, said formulation comprising an amount of active material effective to treat a symptom, condition, and/or disease, wherein said first container is adapted to coupled to a programmable transdermal drug delivery system.
14. The kit of claim 11 wherein the first container includes a specification of an ad¬ ministration schedule that is used to program the programmable transdermal drug delivery system.
15. A method for delivering a drug to a human or animal comprising: a) providing a transdermal drug delivery device coupled to the human or animal, the delivery device having a source of the drug, a programmable timing mechanism, and a mechanism for causing the drug to be delivered transdermally in response to the timing mechanism; and b) timing routines implemented by the timing mechanism, wherein the timing routines are selected to deliver the drug at a time when the human or animal is expected to be asleep.
16. The method of claim 15 wherein the timing routines terminate drug delivery at a time when the human or animal is expected to be asleep.
17. The method of claim 15 wherein the timing routines increase the dosage of drug delivered at a time when the human or animal is expected to be asleep.
18. The method of claim 15 wherein the timing routines decrease the dosage of drug delivered at a time when the human or animal is expected to be asleep
19. The method of claim 15 whereby permeation through the skin is assisted using micro- fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, or a wide range of chemical permeation enhancers.
20. The method of claim 17 whereby permeation through the skin is assisted using micro-fabricated structures commonly referred to as Micro-needles, or heat, or iontophoresis, sonophoresis, or a wide range of chemical permeation enhancers.
PCT/US2005/032672 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery WO2006031856A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA2580329A CA2580329C (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery
EP05796698.8A EP1802258A4 (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery
JP2007531458A JP5254616B2 (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery
AU2005284908A AU2005284908B2 (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US60941804P 2004-09-13 2004-09-13
US60/609,418 2004-09-13
US11/162,525 US7780981B2 (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery
US11/162,517 US20070191815A1 (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery
US11/162,517 2005-09-13

Publications (2)

Publication Number Publication Date
WO2006031856A2 true WO2006031856A2 (en) 2006-03-23
WO2006031856A3 WO2006031856A3 (en) 2006-08-17

Family

ID=38024536

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/032672 WO2006031856A2 (en) 2004-09-13 2005-09-13 Biosynchronous transdermal drug delivery

Country Status (6)

Country Link
US (10) US7780981B2 (en)
EP (1) EP1802258A4 (en)
JP (1) JP5254616B2 (en)
AU (1) AU2005284908B2 (en)
CA (1) CA2580329C (en)
WO (1) WO2006031856A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009076363A1 (en) * 2007-12-10 2009-06-18 Isis Biopolymer Llc Iontophoretic drug delivery device and software application
DE102010030502A1 (en) 2010-06-24 2011-12-29 HSG-IMIT-Institut für Mikro- und Informationstechnologie Quellstoffaktor and conveyor, in particular for a medicinal active substance
WO2013136176A1 (en) * 2012-03-13 2013-09-19 Becton Dickinson France Injection device having a miniaturized drug delivery portion
WO2017019526A2 (en) 2015-07-24 2017-02-02 Kimberly-Clark Worldwide, Inc. Methods for better delivery of active agents to tumors

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100260669A1 (en) * 2004-05-13 2010-10-14 Anthony Joonkyoo Yun Treatment of Seasonal Conditions Through Modulation of the Autonomic Nervous System
US7780981B2 (en) 2004-09-13 2010-08-24 Chrono Therapeutics, Inc. Biosynchronous transdermal drug delivery
US8252321B2 (en) 2004-09-13 2012-08-28 Chrono Therapeutics, Inc. Biosynchronous transdermal drug delivery for longevity, anti-aging, fatigue management, obesity, weight loss, weight management, delivery of nutraceuticals, and the treatment of hyperglycemia, alzheimer's disease, sleep disorders, parkinson's disease, aids, epilepsy, attention deficit disorder, nicotine addiction, cancer, headache and pain control, asthma, angina, hypertension, depression, cold, flu and the like
US7459469B2 (en) 2004-11-10 2008-12-02 Targacept, Inc. Hydroxybenzoate salts of metanicotine compounds
US20070042026A1 (en) * 2005-03-17 2007-02-22 Wille John J Prophylactic and therapeutic treatment of topical and transdermal drug-induced skin reactions
WO2006127905A2 (en) 2005-05-24 2006-11-30 Chrono Therapeutics, Inc. Portable drug delivery device
US8852638B2 (en) 2005-09-30 2014-10-07 Durect Corporation Sustained release small molecule drug formulation
EP2021325A2 (en) * 2006-05-09 2009-02-11 AstraZeneca AB Salt forms of (2s)-(4e)-n-methyl-5-[3-(5-isopropoxypyridin)yl]-4-penten-2-amine
TWI389889B (en) * 2006-05-09 2013-03-21 Targacept Inc Novel polymorph forms of (2s)-(4e)-n-methyl-5-[3-(5-isopropoxypyridin)yl]-4-penten-2-amine
US20090264814A1 (en) * 2006-08-02 2009-10-22 Koninklijke Philips Electronics N.V. Device for medical treatment decision support and/or monitoring the status of a patient
EP2068695A2 (en) * 2006-09-13 2009-06-17 Koninklijke Philips Electronics N.V. Device for automatic adjustment of the dose of melatonin and/or delivery of melatonin
US20080107719A1 (en) * 2006-11-08 2008-05-08 Sukhon Likitlersuang Transdermal drug delivery system
WO2008079868A1 (en) * 2006-12-22 2008-07-03 Drugtech Corporation Clonidine composition and method of use
EP2112923A1 (en) * 2007-01-22 2009-11-04 Targacept Inc. Intranasal, buccal, and sublingual administration of metanicotine analogs
US7715679B2 (en) 2007-05-07 2010-05-11 Adc Telecommunications, Inc. Fiber optic enclosure with external cable spool
JP2010531807A (en) 2007-05-25 2010-09-30 トルマー セラピューティクス, インコーポレイテッド Slow-broadcast formulation of risperidone compound
EP2182932A2 (en) * 2007-07-31 2010-05-12 Targacept Inc. Transdermal administration of (2s)-(4e)-n-methyl-5-(3-(5-isopropoxypyridin)yl)-4-penten-2-amine
EP2178598A4 (en) * 2007-08-17 2012-08-15 Isis Biopolymer Llc Iontophoretic drug delivery system
US20100121307A1 (en) * 2007-08-24 2010-05-13 Microfabrica Inc. Microneedles, Microneedle Arrays, Methods for Making, and Transdermal and/or Intradermal Applications
EP2141620B1 (en) * 2008-07-01 2018-10-31 Roche Diabetes Care GmbH Insulin pump and method for controlling a user interface of an insulin pump
US20100163029A1 (en) 2008-12-30 2010-07-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Method for administering an inhalable compound
US20100169260A1 (en) * 2008-12-30 2010-07-01 Searete Llc Methods and systems for presenting an inhalation experience
EP2419085A4 (en) * 2009-04-14 2013-04-24 Univ California Improved oral drug devices and drug formulations
US20110092881A1 (en) * 2009-05-08 2011-04-21 Isis Biopolymer Inc. Iontophoretic device with contact sensor
US20100286590A1 (en) * 2009-05-08 2010-11-11 Isis Biopolymer Llc Iontophoretic device with improved counterelectrode
CA2769030C (en) 2009-07-30 2016-05-10 Tandem Diabetes Care, Inc. Infusion pump system with disposable cartridge having pressure venting and pressure feedback
RU2012150449A (en) 2010-05-20 2014-06-27 Астразенека Аб A NEW METHOD FOR PRODUCING ARIL SUBSTITUTED OLEFIN AMINES
US9272044B2 (en) 2010-06-08 2016-03-01 Indivior Uk Limited Injectable flowable composition buprenorphine
GB2481017B (en) 2010-06-08 2015-01-07 Rb Pharmaceuticals Ltd Microparticle buprenorphine suspension
US8623409B1 (en) 2010-10-20 2014-01-07 Tris Pharma Inc. Clonidine formulation
WO2013006643A1 (en) 2011-07-06 2013-01-10 The Parkinson's Institute Compositions and methods for treatment of symptoms in parkinson's disease patients
US9539201B2 (en) 2012-04-20 2017-01-10 KAT Transdermals LLC Selegiline transdermal system
US9180242B2 (en) 2012-05-17 2015-11-10 Tandem Diabetes Care, Inc. Methods and devices for multiple fluid transfer
US9265458B2 (en) 2012-12-04 2016-02-23 Sync-Think, Inc. Application of smooth pursuit cognitive testing paradigms to clinical drug development
US20140207047A1 (en) * 2013-01-22 2014-07-24 Chrono Therapeutics, Inc. Transdermal drug delivery system and method
US10105487B2 (en) 2013-01-24 2018-10-23 Chrono Therapeutics Inc. Optimized bio-synchronous bioactive agent delivery system
US9380976B2 (en) 2013-03-11 2016-07-05 Sync-Think, Inc. Optical neuroinformatics
US10201656B2 (en) 2013-03-13 2019-02-12 Tandem Diabetes Care, Inc. Simplified insulin pump for type II diabetics
US9173998B2 (en) 2013-03-14 2015-11-03 Tandem Diabetes Care, Inc. System and method for detecting occlusions in an infusion pump
US9492608B2 (en) 2013-03-15 2016-11-15 Tandem Diabetes Care, Inc. Method and device utilizing insulin delivery protocols
JP6461974B2 (en) * 2013-09-11 2019-01-30 インキューブ ラブズ,リミテッド ライアビリティー カンパニーInCube Labs,LLC System and method for controlling iontophoretic delivery of therapeutic agents based on user inhalation
FR3015300B1 (en) * 2013-12-20 2018-03-02 L'oreal IONTOPHORESE DEVICE WITH TANK
GB201404139D0 (en) 2014-03-10 2014-04-23 Rb Pharmaceuticals Ltd Sustained release buprenorphine solution formulations
US10213586B2 (en) 2015-01-28 2019-02-26 Chrono Therapeutics Inc. Drug delivery methods and systems
US11147955B2 (en) 2015-02-18 2021-10-19 Alma Therapeutics Ltd. Regulator device for drug patch
US10004887B2 (en) 2015-02-18 2018-06-26 Alma Therapeutics Ltd. Transdermal delivery assembly
AU2016228779A1 (en) 2015-03-12 2017-09-07 Chrono Therapeutics Inc. Craving input and support system
DK3283158T3 (en) * 2015-04-17 2023-06-06 Georgia Tech Res Inst Drug delivery device with separable microneedles
WO2017079764A1 (en) * 2015-11-06 2017-05-11 Bkr Ip Holdco Llc Method for the attenuation enhancement of absorbent materials used in both passive and active transdermal drug delivery systems
DK3386563T3 (en) 2015-12-11 2020-01-27 Seraip Ag LIQUID INTERFACE DEVICE FOR DELIVERING LIQUID TO AND / OR TO DRAW LIQUID FROM A PATIENT
EP3416718A4 (en) 2016-02-18 2020-03-11 Alma Therapeuthics Ltd. Regulator device for drug patch
EP3321899A1 (en) * 2016-11-10 2018-05-16 Therasolve NV Electronic communication unit for an adhesive patch
JP2020503950A (en) 2017-01-06 2020-02-06 クロノ セラピューティクス インコーポレイテッドChrono Therapeutics Inc. Device and method for transdermal drug delivery
CA3101966A1 (en) 2018-05-29 2019-12-05 Morningside Venture Investments Limited Drug delivery methods and systems
WO2019241711A1 (en) * 2018-06-14 2019-12-19 Sabiev Anton Wireless iontophoresis patch and controller
US11918689B1 (en) 2020-07-28 2024-03-05 Tris Pharma Inc Liquid clonidine extended release composition
WO2023276090A1 (en) * 2021-06-30 2023-01-05 シンクランド株式会社 Transdermal administration apparatus and transdermal administration system
WO2023108207A1 (en) * 2021-12-17 2023-06-22 Cosmoaesthetics Pty Ltd A wearable transdermal drug applicator

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5538503A (en) * 1993-09-15 1996-07-23 Henley; Julian L. Programmable apparatus for reducing substance dependency in transdermal drug delivery

Family Cites Families (381)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2183482A (en) 1936-12-23 1939-12-12 Schmid Inc Julius Injector
US3279653A (en) 1964-12-17 1966-10-18 Frederick W Pfleger Escapement controlled dispensing apparatus
US3845217A (en) 1972-11-16 1974-10-29 Helsingborg L Ab Buffered smoking substitute compositions
GB1528391A (en) 1976-01-05 1978-10-11 Gildemeister V Aerosol compositions
GB2030862A (en) 1978-04-10 1980-04-16 Smith A A composition for treating drug addiction such as smoking nicotine-containing products
US4321387A (en) 1980-03-21 1982-03-23 Philip Morris, Incorporated Process for the preparation of optically active nicotine analogs
US4313439A (en) 1980-03-24 1982-02-02 Biotek, Inc. Automated, spring-powered medicament infusion system
US4327725A (en) 1980-11-25 1982-05-04 Alza Corporation Osmotic device with hydrogel driving member
US4590278A (en) 1981-01-29 1986-05-20 Philip Morris Incorporated Nicotine analogs
US4332945A (en) 1981-01-29 1982-06-01 Philip Morris, Incorporated Optically active nicotine analogs and process for their preparation
US4379454A (en) * 1981-02-17 1983-04-12 Alza Corporation Dosage for coadministering drug and percutaneous absorption enhancer
LU84485A1 (en) * 1982-11-22 1984-06-13 Oreal NOVEL ANTI-ACNE COMPOSITION BASED ON BENZOYL PEROXIDE AND AT LEAST ONE OTHER ACTIVE INGREDIENT
GB8301659D0 (en) 1983-01-21 1983-02-23 Leo Ab Smoking substitutes
US4856188A (en) 1984-10-12 1989-08-15 Drug Delivery Systems Inc. Method for making disposable and/or replenishable transdermal drug applicators
GB8317576D0 (en) 1983-06-29 1983-08-03 Shaw A S W Consumer tobacco products
GB2142822B (en) 1983-06-29 1987-02-11 Alec Stanley Walter Shaw Nicotine lozenges
US5865786A (en) 1983-08-18 1999-02-02 Drug Delivery Systems, Inc. Programmable control and mounting system for transdermal drug applicator
US4708716A (en) 1983-08-18 1987-11-24 Drug Delivery Systems Inc. Transdermal drug applicator
BE899037A (en) 1984-02-29 1984-06-18 Trenker Ady Nicotine compsn. to help people to stop smoking - is a tablet for sucking, which contains nicotine or its water-soluble acid salt
US4920989A (en) 1985-04-25 1990-05-01 Regents Of The University Of California Method and apparatus for aiding in the reduction of incidence of tobacco smoking
US4953572A (en) 1985-04-25 1990-09-04 Rose Jed E Method and apparatus for aiding in the reduction of incidence of tobacco smoking
DE3586595T2 (en) 1985-06-10 1993-04-08 Drug Delivery Systems Inc PROGRAMMABLE CONTROL AND BUILDING SYSTEM OF AN ADMINISTRATOR FOR TRANSDERMIC MEDICINES.
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US4853854A (en) 1985-12-26 1989-08-01 Health Innovations, Inc. Human behavior modification which establishes and generates a user adaptive withdrawal schedule
US4772263A (en) 1986-02-03 1988-09-20 Regents Of The University Of Minnesota Spring driven infusion pump
DE3645036A1 (en) 1986-11-18 1989-01-05 Forschungsgesellschaft Rauchen Nicotine-containing composition
US4917676A (en) 1986-11-20 1990-04-17 Ciba-Geigy Corporation User-activated transdermal therapeutic system
AT391998B (en) 1987-02-02 1990-12-27 Falko Dr Skrabal Device for determining the concentration of at least one medicinal substance in living organisms
US5049387A (en) 1987-03-09 1991-09-17 Alza Corporation Inducing skin tolerance to a sensitizing drug
US5000956A (en) 1987-03-09 1991-03-19 Alza Corporation Prevention of contact allergy by coadministration of a corticosteroid with a sensitizing drug
US5013293A (en) 1987-05-28 1991-05-07 Drug Delivery Systems Inc. Pulsating transdermal drug delivery system
US5001139A (en) 1987-06-12 1991-03-19 American Cyanamid Company Enchancers for the transdermal flux of nivadipine
US4992445A (en) 1987-06-12 1991-02-12 American Cyanamid Co. Transdermal delivery of pharmaceuticals
US5388571A (en) 1987-07-17 1995-02-14 Roberts; Josephine A. Positive-pressure ventilator system with controlled access for nebulizer component servicing
US5073380A (en) 1987-07-27 1991-12-17 Mcneil-Ppc, Inc. Oral sustained release pharmaceutical formulation and process
EP0311313B1 (en) 1987-10-05 1995-05-10 Yamanouchi Pharmaceutical Co. Ltd. Heterocyclic spiro compounds and their preparation
JPH01265021A (en) 1987-10-29 1989-10-23 Hercon Lab Corp Article for discharging and supplying composition containing pharmacologically active substance to animal tissue in controllable manner
US4917895A (en) * 1987-11-02 1990-04-17 Alza Corporation Transdermal drug delivery device
US4885154A (en) 1988-03-01 1989-12-05 Alza Corporation Method for reducing sensitization or irritation in transdermal drug delivery and means therefor
US5472946A (en) 1988-04-08 1995-12-05 Peck; James V. Transdermal penetration enhancers
US5364630A (en) 1988-06-14 1994-11-15 Alza Corporation Subsaturated nicotine transdermal therapeutic system
US5028428A (en) 1988-08-09 1991-07-02 Estee Lauder Inc. Anti-irritant and desensitizing compositions and methods of their use
US4952928A (en) 1988-08-29 1990-08-28 B. I. Incorporated Adaptable electronic monitoring and identification system
US5496266A (en) 1990-04-30 1996-03-05 Alza Corporation Device and method of iontophoretic drug delivery
US5236714A (en) 1988-11-01 1993-08-17 Alza Corporation Abusable substance dosage form having reduced abuse potential
US5248501A (en) 1988-11-22 1993-09-28 Parnell Pharmaceuticals Drug delivery systems containing eriodictyon fluid extract as an excipient, and methods and compositions associated therewith
IE62662B1 (en) 1989-01-06 1995-02-22 Elan Corp Plc Use of nicotine in the treatment of conditions susceptible to said treatment
JP2638635B2 (en) 1989-01-31 1997-08-06 積水化学工業株式会社 Transdermal formulation
JPH02208813A (en) 1989-02-09 1990-08-20 Fuji Photo Film Co Ltd Thin film magnetic head
US4908213A (en) 1989-02-21 1990-03-13 Schering Corporation Transdermal delivery of nicotine
GB2230439A (en) 1989-04-20 1990-10-24 Alec Stanley Walter Shaw Nicotine lozenges
US5262165A (en) 1992-02-04 1993-11-16 Schering Corporation Transdermal nitroglycerin patch with penetration enhancers
US5525351A (en) 1989-11-07 1996-06-11 Dam; Anders Nicotine containing stimulant unit
US5512306A (en) 1992-06-19 1996-04-30 Pharmica Ab Smoking substitute
US5149719A (en) 1990-04-27 1992-09-22 Minnesota Mining And Manufacturing Company Composition for transdermal penetration of medicaments
US5130139A (en) 1990-07-06 1992-07-14 Alza Corporation Reduction or prevention of skin irritation by drugs
US5120545A (en) 1990-08-03 1992-06-09 Alza Corporation Reduction or prevention of sensitization to drugs
US5242941A (en) * 1990-12-04 1993-09-07 State Of Oregon Methods of treating circadian rhythm disorders
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
US5232704A (en) 1990-12-19 1993-08-03 G. D. Searle & Co. Sustained release, bilayer buoyant dosage form
US5221254A (en) 1991-04-02 1993-06-22 Alza Corporation Method for reducing sensation in iontophoretic drug delivery
JP3153551B2 (en) 1991-05-29 2001-04-09 アボツト・ラボラトリーズ Isoxazole and isothiazole compounds that enhance cognitive function
US5149538A (en) 1991-06-14 1992-09-22 Warner-Lambert Company Misuse-resistive transdermal opioid dosage form
US5464387A (en) 1991-07-24 1995-11-07 Alza Corporation Transdermal delivery device
US5273755A (en) 1991-08-23 1993-12-28 Cygnus Therapeutic Systems Transdermal drug delivery device using a polymer-filled microporous membrane to achieve delayed onset
US5273756A (en) 1991-08-23 1993-12-28 Cygnus Therapeutic Systems Transdermal drug delivery device using a membrane-protected microporous membrane to achieve delayed onset
US5352456A (en) * 1991-10-10 1994-10-04 Cygnus Therapeutic Systems Device for administering drug transdermally which provides an initial pulse of drug
DE4138352C2 (en) * 1991-11-21 2000-03-16 Stuertz Maschbau Method and device for producing rectangular frames
US5304739A (en) 1991-12-19 1994-04-19 Klug Reja B High energy coaxial cable for use in pulsed high energy systems
GB9200047D0 (en) 1992-01-03 1992-02-26 Univ Alberta Nicotine-containing nasal spray
US6436078B1 (en) 1994-12-06 2002-08-20 Pal Svedman Transdermal perfusion of fluids
ATE192646T1 (en) * 1992-01-21 2000-05-15 Howard Alliger USE OF A MIXTURE OF SUBSTANCES CONTAINING ALPHA HYDROXY ORGANIC ACID FOR THE PRODUCTION OF A MEDICINAL PRODUCT FOR THE TREATMENT OF MINOR ORAL ULCERS
IL104365A0 (en) 1992-01-31 1993-05-13 Gensia Pharma Method and apparatus for closed loop drug delivery
CH686816A5 (en) 1992-02-04 1996-07-15 Asulab Ag Means for delivering a drug.
US5242934A (en) 1992-03-02 1993-09-07 R. J. Reynolds Tobacco Company Method for treatment of neurodegenerative diseases
US5212188A (en) 1992-03-02 1993-05-18 R. J. Reynolds Tabacco Company Method for treatment of neurodegenerative diseases
US5242935A (en) 1992-03-06 1993-09-07 R. J. Reynolds Tobacco Company Method for treatment of neurodegenerative diseases
US5405614A (en) 1992-04-08 1995-04-11 International Medical Associates, Inc. Electronic transdermal drug delivery system
US5756117A (en) * 1992-04-08 1998-05-26 International Medical Asscociates, Inc. Multidose transdermal drug delivery system
US5227391A (en) 1992-04-10 1993-07-13 R. J. Reynolds Tobacco Company Method for treatment of neurodegenerative diseases
US5232933A (en) 1992-05-21 1993-08-03 R. J. Reynolds Tobacco Company Method for treatment of neurodegenerative diseases
US5310404A (en) 1992-06-01 1994-05-10 Alza Corporation Iontophoretic delivery device and method of hydrating same
US5501697A (en) 1992-06-23 1996-03-26 Fisher; Gary R. Treatment device to aid in long-term cessation of smoking
CA2072040C (en) 1992-06-23 2002-06-11 Charles Borg Method and device to facilitate the cognitive development of alternative response behaviour
US5248690A (en) 1992-07-07 1993-09-28 R. J. Reynolds Tobacco Company Method for treatment of neurodegenerative diseases
US5252604A (en) 1992-07-10 1993-10-12 Hoffmann-La Roche Inc. Compositions of retinoic acids and tocopherol for prevention of dermatitis
EP0656771A4 (en) 1992-08-25 1996-07-31 Cygnus Therapeutic Systems Printed transdermal drug delivery device.
IL107184A (en) 1992-10-09 1997-08-14 Abbott Lab Heterocyclic ether compounds that enhance cognitive function
AU5551394A (en) 1992-11-09 1994-06-08 Pharmetrix Corporation Combined analgesic delivery methods for pain management
CA2114968A1 (en) 1993-02-25 1994-08-26 John Wille Transdermal treatment with mast cell degranulating agents for drug-induced hypersensitivity
US5843979A (en) 1993-02-25 1998-12-01 Bristol-Myers Squibb Company Transdermal treatment with mast cell degranulating agents for drug-induced hypersensitivity
US5516793A (en) 1993-04-26 1996-05-14 Avon Products, Inc. Use of ascorbic acid to reduce irritation of topically applied active ingredients
US5445609A (en) 1993-05-28 1995-08-29 Alza Corporation Electrotransport agent delivery device having a disposable component and a removable liner
US5451407A (en) 1993-06-21 1995-09-19 Alza Corporation Reduction or prevention of skin irritation or sensitization during transdermal administration of a irritating or sensitizing drug
WO1995003028A1 (en) * 1993-07-23 1995-02-02 Morris Herstein Cosmetic, skin-renewal stimulating composition with long-term irritation control
US5549906A (en) 1993-07-26 1996-08-27 Pharmacia Ab Nicotine lozenge and therapeutic method for smoking cessation
US5377258A (en) 1993-08-30 1994-12-27 National Medical Research Council Method and apparatus for an automated and interactive behavioral guidance system
US5533971A (en) 1993-09-03 1996-07-09 Alza Corporation Reduction of skin irritation during electrotransport
AU662877B3 (en) 1993-10-21 1995-09-14 Amal Hakim Khalil Nicotine lozenges
US5997501A (en) 1993-11-18 1999-12-07 Elan Corporation, Plc Intradermal drug delivery device
DE4400770C1 (en) 1994-01-13 1995-02-02 Lohmann Therapie Syst Lts Plaster containing an active substance for delivery of oestradiol with at least one penetration enhancer, method of producing it and its use
US5393526A (en) 1994-02-07 1995-02-28 Elizabeth Arden Company, Division Of Conopco, Inc. Cosmetic compositions
CA2187023A1 (en) * 1994-04-13 1995-10-26 Steven Andrew Giannos Temporally controlled drug delivery systems
US6121276A (en) 1994-04-22 2000-09-19 Pentech Pharmaceuticals, Inc. Apomorphine-containing dosage forms for ameliorating male erectile dysfunction
US5876368A (en) 1994-09-30 1999-03-02 Becton Dickinson And Company Iontophoretic drug delivery device having improved controller
US5445823A (en) 1994-10-20 1995-08-29 The Procter & Gamble Company Dermatological compositions and method of treatment of skin lesions therewith
US5505958A (en) * 1994-10-31 1996-04-09 Algos Pharmaceutical Corporation Transdermal drug delivery device and method for its manufacture
US5703100A (en) 1994-11-10 1997-12-30 Sibia Neurosciences, Inc. Modulators of acetylcholine receptors
US5686100A (en) 1994-11-22 1997-11-11 E.R. Squibb & Sons, Inc. Prophylactic and therapeutic treatment of skin sensitization and irritation
US5618557A (en) 1994-11-22 1997-04-08 E.R. Squibb & Sons, Inc. Prophylactic treatment of allergic contact dermatitis
US5697896A (en) 1994-12-08 1997-12-16 Alza Corporation Electrotransport delivery device
DE4444238A1 (en) 1994-12-13 1996-06-20 Beiersdorf Ag Cosmetic or dermatological drug combinations of cinnamic acid derivatives and flavone glycosides
FR2728793A1 (en) 1994-12-28 1996-07-05 Oreal Compsns. for treating sensitive skin
US5562607A (en) 1995-01-18 1996-10-08 Alza Corporation Electrotransport device having reusable controller power saver
US5879322A (en) 1995-03-24 1999-03-09 Alza Corporation Self-contained transdermal drug delivery device
WO1996032142A1 (en) 1995-04-12 1996-10-17 Hopp Robert B Skin patch for use in contact immunotherapy
US5601839A (en) 1995-04-26 1997-02-11 Theratech, Inc. Triacetin as a penetration enhancer for transdermal delivery of a basic drug
US5882676A (en) 1995-05-26 1999-03-16 Alza Corporation Skin permeation enhancer compositions using acyl lactylates
IL118279A (en) 1995-06-07 2006-10-05 Abbott Lab 3 - pyridyloxy (or thio) alkyl heterocyclic compounds, pharmaceutical compositions containing them and their uses in the preparation of medicaments for controlling chemical synaptic transmission
US6093419A (en) 1995-06-07 2000-07-25 Lectec Corporation Compliance verification method and device in compulsory drug administration
US6245347B1 (en) 1995-07-28 2001-06-12 Zars, Inc. Methods and apparatus for improved administration of pharmaceutically active compounds
US5936088A (en) 1995-09-22 1999-08-10 Novo Nordisk A/S Substituted azacyclic or azabicyclic compounds with affinity and selectivity for nicotinic cholinergic receptors
AU7083596A (en) 1995-09-22 1997-04-09 Novo Nordisk A/S Novel substituted azacyclic or azabicyclic compounds
US5688232A (en) 1995-09-28 1997-11-18 Becton Dickinson And Company Iontophoretic drug delivery device having an improved controller
CA2233197A1 (en) 1995-09-28 1997-04-03 Becton, Dickinson And Company Iontophoretic drug delivery system, including reusable device
DE69628333T2 (en) 1995-11-17 2004-04-08 Merck & Co., Inc. NEW SUBSTITUTED ARYL COMPOUNDS AS ACETYLCHOLINE RECEPTORS MODULATORS
US8092224B2 (en) 1995-11-22 2012-01-10 James A. Jorasch Systems and methods for improved health care compliance
AU1058797A (en) 1995-11-22 1997-06-11 Bristol-Myers Squibb Company Treatment with calcium channel blockers for drug-induced hypersensitivity
US5858001A (en) 1995-12-11 1999-01-12 Elan Medical Technologies Limited Cartridge-based drug delivery device
US5846983A (en) 1996-02-09 1998-12-08 Mayo Foundation For Medical Education And Research Colonic delivery of nicotine to treat inflammatory bowel disease
US5733269A (en) 1996-03-15 1998-03-31 Fuisz Technologies Ltd. Method and kit for positioning transdermal delivery system
WO1997034605A1 (en) 1996-03-21 1997-09-25 Mayo Foundation For Medical Education And Research Use of nicotine to treat liver disease
US6271240B1 (en) 1996-05-06 2001-08-07 David Lew Simon Methods for improved regulation of endogenous dopamine in prolonged treatment of opioid addicted individuals
US5785688A (en) 1996-05-07 1998-07-28 Ceramatec, Inc. Fluid delivery apparatus and method
JP3707798B2 (en) 1996-05-13 2005-10-19 ノバルティス・コンシューマー・ヘルス・ソシエテ・アノニム Intraoral drug delivery system
US5629325A (en) 1996-06-06 1997-05-13 Abbott Laboratories 3-pyridyloxymethyl heterocyclic ether compounds useful in controlling chemical synaptic transmission
US5716987A (en) 1996-06-21 1998-02-10 Bristol-Myers Squibb Company Prophylactic and therapeutic treatment of skin sensitization and irritation
GB9614902D0 (en) 1996-07-16 1996-09-04 Rhodes John Sustained release composition
US5788671A (en) 1996-08-14 1998-08-04 Sims Deltec, Inc. Reusable cassette housings and methods
US6081734A (en) 1996-08-16 2000-06-27 Roche Diagnostics Gmbh Monitoring system for the regular intake of a medicament
US5972389A (en) 1996-09-19 1999-10-26 Depomed, Inc. Gastric-retentive, oral drug dosage forms for the controlled-release of sparingly soluble drugs and insoluble matter
US5797867A (en) 1996-09-27 1998-08-25 Becton Dickinson And Company Iontophoretic drug delivery system, including method for activating same for attachment to patient
US5919156A (en) 1996-09-27 1999-07-06 Becton, Dickinson And Company Iontophoretic drug delivery system, including unit for dispensing patches
US6746688B1 (en) 1996-10-13 2004-06-08 Neuroderm Ltd. Apparatus for the transdermal treatment of Parkinson's disease
US5800832A (en) 1996-10-18 1998-09-01 Virotex Corporation Bioerodable film for delivery of pharmaceutical compounds to mucosal surfaces
HU222607B1 (en) 1996-11-21 2003-08-28 Laboratoires D'hygiene Et De Dietetique (L.H.D.) Miniature valve, device for filling the reservoir, apparatus for the transdermal administration of medicine, method of manufacturing said valve
DE19650115C1 (en) 1996-12-03 1998-07-02 Fraunhofer Ges Forschung Medication dosing device
US5908301A (en) 1996-12-05 1999-06-01 Lutz; Raymond Method and device for modifying behavior
US8734339B2 (en) 1996-12-16 2014-05-27 Ip Holdings, Inc. Electronic skin patch for real time monitoring of cardiac activity and personal health management
US6019997A (en) 1997-01-09 2000-02-01 Minnesota Mining And Manufacturing Hydroalcoholic compositions for transdermal penetration of pharmaceutical agents
US6018679A (en) 1997-01-29 2000-01-25 Novartis Finance Corp. Iontophoretic transdermal delivery and control of adverse side-effects
US6020335A (en) 1997-02-06 2000-02-01 Pfizer Inc (N-(pyridinylmethyl)-heterocyclic)ylideneamine compounds as nicotinic acetylcholine receptor binding agents
US5860957A (en) 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US5861423A (en) 1997-02-21 1999-01-19 Caldwell; William Scott Pharmaceutical compositions incorporating aryl substituted olefinic amine compounds
HUP0000759A3 (en) 1997-03-11 2000-11-28 Darwin Discovery Ltd Cambridge Dosage forms comprising separate portions of r- and s-enantiomers
FR2761072B1 (en) 1997-03-20 1999-04-23 Synthelabo 2,3-DIHYDROFURO [3,2-B] PYRIDINE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
US5977131A (en) 1997-04-09 1999-11-02 Pfizer Inc. Azaindole-ethylamine derivatives as nicotinic acetylcholine receptor binding agents
WO1998046093A1 (en) 1997-04-14 1998-10-22 Robert Lindburg Smoking control device
US6024981A (en) 1997-04-16 2000-02-15 Cima Labs Inc. Rapidly dissolving robust dosage form
NZ500643A (en) 1997-05-30 2001-12-21 Neurosearch As Spiro-quinuclidine derivatives and their use in treating conditions responsive to nicotinic ACh receptor modulators
AU736547B2 (en) 1997-05-30 2001-08-02 Neurosearch A/S Azacyclooctane and heptane derivatives, their preparation and use in therapy
AU736798B2 (en) 1997-05-30 2001-08-02 Neurosearch A/S 9-Azabicyclo(3.3.1)non-2-ene and nonane derivatives as cholinergic ligands at nicotinic ACh receptors
DK0984965T3 (en) 1997-05-30 2004-09-27 Neurosearch As 8-azabicyclo [3.2.1] oct-2 end derivatives as cholinergic ligands at nicotine ACh receptors
EP0998271B3 (en) 1997-06-06 2014-10-29 Depomed, Inc. Gastric-retentive oral drug dosage forms for controlled release of highly soluble drugs
FR2765874B1 (en) 1997-07-09 1999-08-13 Synthelabo 6-PYRROLIDIN-2-YLPYRINDINE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
AR013184A1 (en) 1997-07-18 2000-12-13 Astrazeneca Ab SPYROZOBICYCLIC HETERO CYCLIC AMINES, PHARMACEUTICAL COMPOSITION, USE OF SUCH AMINES TO PREPARE MEDICINES AND METHOD OF TREATMENT OR PROPHYLAXIS
US6420622B1 (en) 1997-08-01 2002-07-16 3M Innovative Properties Company Medical article having fluid control film
US6034079A (en) 1997-08-11 2000-03-07 University Of South Florida Nicotine antagonists for nicotine-responsive neuropsychiatric disorders
PT1003476E (en) 1997-08-11 2005-05-31 Alza Corp ACTIVE AGGREGATE AGGREGATE DOSAGE FORM ADAPTED FOR GASTRIC RETENTION
DE19738855C2 (en) 1997-09-05 2001-01-04 Lohmann Therapie Syst Lts Transdermal therapeutic system with adhesive reservoir layer and unidirectional elastic back layer
US5786888A (en) * 1997-09-16 1998-07-28 Trw Inc. Pulsed radiation classifier and related method
US6660295B2 (en) 1997-09-30 2003-12-09 Alza Corporation Transdermal drug delivery device package with improved drug stability
US7011843B2 (en) 1997-10-01 2006-03-14 Lts Lohmann-Therapie Systeme Ag Method for protecting a human being against health impairment by ingestion of a transdermal therapeutic system
US5879292A (en) 1997-10-09 1999-03-09 Edward A. Sternberg Bandage including data acquisition components
WO1999021834A1 (en) 1997-10-27 1999-05-06 Neurosearch A/S Heteroaryl diazacycloalkanes as cholinergic ligands at nicotinic acetylcholine receptors
US6162214A (en) 1997-10-30 2000-12-19 Eclipse Surgical Technologies, Inc. Corning device for myocardial revascularization
EP1357121A1 (en) 1997-11-05 2003-10-29 Neurosearch A/S Azaring-ether deritatives and their use as nicotinic ach receptor modulators
DK1510213T3 (en) 1997-11-10 2009-03-23 Strakan Int Ltd Penetration-enhancing and irritation-reducing systems including testosterone
US6374136B1 (en) * 1997-12-22 2002-04-16 Alza Corporation Anhydrous drug reservoir for electrolytic transdermal delivery device
CA2315890C (en) 1997-12-22 2009-08-11 Alza Corporation Rate controlling membranes for controlled drug delivery devices
US6059736A (en) 1998-02-24 2000-05-09 Tapper; Robert Sensor controlled analysis and therapeutic delivery system
CA2231968A1 (en) 1998-03-11 1999-09-11 Smoke-Stop, A Partnership Consisting Of Art Slutsky Method of producing a nicotine medicament
JP4425465B2 (en) 1998-03-23 2010-03-03 エラン コーポレーション ピーエルシー Drug delivery device
US5945123A (en) * 1998-04-02 1999-08-31 K-V Pharmaceutical Company Maximizing effectiveness of substances used to improve health and well being
US6057446A (en) 1998-04-02 2000-05-02 Crooks; Peter Anthony Certain 1-aza-tricyclo [3.3.1-13,7 ] decane compounds
US20040062802A1 (en) 1998-04-02 2004-04-01 Hermelin Victor M. Maximizing effectiveness of substances used to improve health and well being
EP0955301A3 (en) 1998-04-27 2001-04-18 Pfizer Products Inc. 7-aza-bicyclo[2.2.1]-heptane derivatives, their preparation and use according to their affinity for neuronal nicotinic acetylcholine receptors
US6211194B1 (en) 1998-04-30 2001-04-03 Duke University Solution containing nicotine
US6232316B1 (en) 1998-06-16 2001-05-15 Targacept, Inc. Methods for treatment of CNS disorders
US5967789A (en) 1998-07-30 1999-10-19 Smoke Stoppers International, Inc. Method and system for stopping or modifying undesirable health-related behavior habits or maintaining desirable health-related behavior habits
US6365182B1 (en) 1998-08-12 2002-04-02 Cima Labs Inc. Organoleptically pleasant in-mouth rapidly disintegrable potassium chloride tablet
US6368625B1 (en) 1998-08-12 2002-04-09 Cima Labs Inc. Orally disintegrable tablet forming a viscous slurry
EP1107965B1 (en) 1998-08-25 2004-08-11 Ortho-McNeil Pharmaceutical, Inc. Pyridyl ethers and thioethers as ligands for nicotinic acetylcholine receptor and its therapeutic application
US6165165A (en) * 1998-10-02 2000-12-26 Genx International, Inc. Embryo-implanting catheter assembly and method for making the same
US6262124B1 (en) 1998-10-22 2001-07-17 Gary Maurice Dull Pharmaceutical compositions and methods for use
US6211296B1 (en) * 1998-11-05 2001-04-03 The B. F. Goodrich Company Hydrogels containing substances
CA2342621A1 (en) 1998-11-27 2000-06-08 Neurosearch A/S 8-azabicyclo[3.2.1]oct-2-ene and -octane derivatives
FR2786769B1 (en) 1998-12-04 2002-10-25 Synthelabo 2,5-DIAZABICYCLO [2.2.1] HEPTANE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
FR2786770B1 (en) 1998-12-04 2001-01-19 Synthelabo NONANE 1,4-DIAZABICYCLO [3.2.2.] DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
ATE320711T1 (en) 1998-12-16 2006-04-15 Univ South Florida EXO-S-MECAMYLAMIN FORMULATION AND ITS USE IN TREATMENTS
KR100383252B1 (en) 1998-12-17 2003-07-16 주식회사 삼양사 Transdermal Dosage Compositions Containing Buprenoline and Patches Comprising the Same
ES2209825T3 (en) 1999-01-29 2004-07-01 Abbott Laboratories USEFUL DIAZABICICLIC DERIVATIVES AS LIGANDS OF THE NICOTINIC RECEIVER OF ACETILCOLINE.
US6583160B2 (en) 1999-04-14 2003-06-24 Steve Smith Nicotine therapy method and oral carrier for assuaging tobacco-addiction
US6416471B1 (en) 1999-04-15 2002-07-09 Nexan Limited Portable remote patient telemonitoring system
US6454708B1 (en) 1999-04-15 2002-09-24 Nexan Limited Portable remote patient telemonitoring system using a memory card or smart card
WO2000064885A1 (en) 1999-04-26 2000-11-02 Neurosearch A/S Heteroaryl diazacycloalkanes, their preparation and use
AU3902300A (en) 1999-04-29 2000-11-17 Dow Chemical Company, The Bis(n,n-dihydrocarbylamino)- substituted cyclopentadienes and metal complexes thereof
US6689380B1 (en) 1999-05-17 2004-02-10 Kevin S. Marchitto Remote and local controlled delivery of pharmaceutical compounds using electromagnetic energy
DE60044084D1 (en) 1999-06-04 2010-05-12 Georgia Tech Res Inst DEVICES FOR THE ENLARGED PENETRATION OF MICRONEDES IN BIOLOGICAL HARDENING
DE19925613A1 (en) * 1999-06-04 2000-12-07 Lohmann Therapie Syst Lts Composite laminate and process for its manufacture
US6567785B2 (en) 1999-06-19 2003-05-20 John Richard Clendenon Electronic behavior modification reminder system and method
DE19958554C2 (en) 1999-07-02 2002-06-13 Lohmann Therapie Syst Lts Microreservoir system based on polysiloxanes and ambiphilic solvents and their manufacture
US20040229908A1 (en) 1999-07-13 2004-11-18 Jodi Nelson Compositions and methods for the treatment of Parkinson's disease and tardive dyskinesias
US20080138399A1 (en) 1999-07-16 2008-06-12 Aradigm Corporation Dual release nicotine formulations, and systems and methods for their use
EP1218048A4 (en) 1999-07-16 2009-08-26 Aradigm Corp System for effecting smoke cessation
US6799576B2 (en) 1999-07-16 2004-10-05 Aradigm Corporation System for effecting smoking cessation
US8256433B2 (en) 1999-07-16 2012-09-04 Aradigm Corporation Systems and methods for effecting cessation of tobacco use
US20080138294A1 (en) 1999-07-16 2008-06-12 Igor Gonda Systems and methods for effecting cessation of tobacco use
US20080138398A1 (en) 1999-07-16 2008-06-12 Aradigm Corporation Dual release nicotine formulations, and systems and methods for their use
US8188043B2 (en) 1999-07-28 2012-05-29 The Board Of Trustees Of The Leland Stanford Jr. University Nicotine in therapeutic angiogenesis and vasculogenesis
CA2381951A1 (en) 1999-08-18 2001-02-22 Microchips, Inc. Thermally-activated microchip chemical delivery devices
US7376700B1 (en) 1999-08-23 2008-05-20 Wellcoaches Corporation Personal coaching system for clients with ongoing concerns such as weight loss
FR2798065B1 (en) 1999-09-02 2003-09-05 Assist Publ Hopitaux De Paris USE OF NICOTINE OR DERIVATIVES THEREOF IN A MEDICAMENT FOR THE TREATMENT OF NEUROLOGICAL DISEASES, ESPECIALLY PARKINSON'S DISEASE
KR20010036685A (en) 1999-10-11 2001-05-07 김윤 Transdermal fentanyl delivery matrix system
SE9903998D0 (en) 1999-11-03 1999-11-03 Astra Ab New compounds
AU1713501A (en) 1999-11-23 2001-06-04 Robert Gordon University, The Bilayered buccal tablets comprising nicotine
US6539250B1 (en) * 1999-12-15 2003-03-25 David S. Bettinger Programmable transdermal therapeutic apparatus
US20020004065A1 (en) * 2000-01-20 2002-01-10 David Kanios Compositions and methods to effect the release profile in the transdermal administration of active agents
US6261595B1 (en) 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
JP2001261652A (en) 2000-03-21 2001-09-26 Suntory Ltd Disubstituted iminoheterocyclic compound
US6437004B1 (en) 2000-04-06 2002-08-20 Nicholas V. Perricone Treatment of skin damage using olive oil polyphenols
US6489025B2 (en) 2000-04-12 2002-12-03 Showa Denko K.K. Fine carbon fiber, method for producing the same and electrically conducting material comprising the fine carbon fiber
US6955821B2 (en) 2000-04-28 2005-10-18 Adams Laboratories, Inc. Sustained release formulations of guaifenesin and additional drug ingredients
AU2001278849A1 (en) 2000-05-19 2001-12-03 Sabinsa Corporation Method of increased bioavailability of nutrients and pharmaceutical preparationswith tetrahydropiperine and its analogues and derivatives
CA2409826C (en) * 2000-06-01 2010-02-02 Science Applications International Corporation Systems and methods for monitoring health and delivering drugs transdermally
BRPI0414359A (en) 2000-06-16 2006-11-14 Bodymedia Inc body weight monitoring and management system and other psychological conditions that include interactive and personalized planning, intervention and reporting
WO2002002081A1 (en) 2000-07-05 2002-01-10 Capricorn Pharma, Inc. Rapid-melt semi-solid compositions, methods of making same and methods of using same
US6589229B1 (en) 2000-07-31 2003-07-08 Becton, Dickinson And Company Wearable, self-contained drug infusion device
JP2002092180A (en) 2000-09-19 2002-03-29 Hiroko Takahashi System for supporting abstention from smoking
AU2001292185A1 (en) 2000-10-12 2002-04-22 Beecham Pharmaceuticals (Pte) Limited Formulation containing amoxicillin
GB0026137D0 (en) 2000-10-25 2000-12-13 Euro Celtique Sa Transdermal dosage form
US6569449B1 (en) 2000-11-13 2003-05-27 University Of Kentucky Research Foundation Transdermal delivery of opioid antagonist prodrugs
US6682757B1 (en) 2000-11-16 2004-01-27 Euro-Celtique, S.A. Titratable dosage transdermal delivery system
DE10102817B4 (en) 2001-01-23 2006-01-12 Lts Lohmann Therapie-Systeme Ag Device and method for heat pulse-assisted transdermal application of active substances
DE10103158A1 (en) 2001-01-24 2002-08-01 Christian Von Falkenhausen Device for electricity-assisted introduction of active agents into the skin comprises at least two electrodes which are connected through galvanic conduction of the reservoir of the active agent
DE10105759C1 (en) 2001-02-08 2001-10-25 Lohmann Therapie Syst Lts Electrically and magnetically assisted charge carrier delivery device e.g. for transdermal application of pharmaceuticals, has electrical and magnetic fields applied to charge carrier reservoir layer
CN1556716A (en) 2001-02-22 2004-12-22 ���Ͽع����޹�˾ Modular infusion device and method
AU2002255627B2 (en) 2001-02-27 2008-01-17 The Regents Of The University Of Michigan Use of natural EGFR inhibitors to prevent side effects due to retinoid therapy, soaps, and other stimuli that activate the epidermal growth receptor
US20020127256A1 (en) * 2001-03-01 2002-09-12 Howard Murad Compositions and methods for treating dermatological disorders
FR2822377A1 (en) 2001-03-23 2002-09-27 Oreal USE OF FIBERS AS ANTI-IRRITANT AGENT IN A COSMETIC OR DERMATOLOGICAL COMPOSITION
CN1553771A (en) 2001-03-26 2004-12-08 史密丝克莱恩比彻姆公司 Nicotine-containing oral dosage form
WO2002076401A2 (en) 2001-03-26 2002-10-03 Dana-Farber Cancer Institute, Inc. Method of attenuating reactions to skin irritants
CA2381630A1 (en) 2001-04-23 2002-10-23 Leonard Theodore Meltzer Method for preventing dyskinesias
ES2318010T3 (en) 2001-05-01 2009-05-01 Euro-Celtique S.A. TRANSDERMAL SYSTEMS CONTAINING OPISONS RESISTANT TO BAD USES.
US6723086B2 (en) * 2001-05-07 2004-04-20 Logiq Wireless Solutions, Inc. Remote controlled transdermal medication delivery device
US20040019321A1 (en) 2001-05-29 2004-01-29 Sage Burton H. Compensating drug delivery system
SE0102197D0 (en) 2001-06-20 2001-06-20 Pharmacia Ab New product and use and manufacture thereof
US6860731B2 (en) * 2001-07-09 2005-03-01 Asm Technology Singapore Pte Ltd. Mold for encapsulating a semiconductor chip
US7332182B2 (en) 2001-08-06 2008-02-19 Purdue Pharma L.P. Pharmaceutical formulation containing opioid agonist, opioid antagonist and irritant
US20030044458A1 (en) 2001-08-06 2003-03-06 Curtis Wright Oral dosage form comprising a therapeutic agent and an adverse-effect agent
US6576269B1 (en) 2001-09-06 2003-06-10 Alexander Y. Korneyev Treating open skin lesions using extract of sea buckthorn
GB0121628D0 (en) 2001-09-07 2001-10-31 Watmough David J Improved method and apparatus for transdermal drug delivery
FR2829644A1 (en) 2001-09-10 2003-03-14 St Microelectronics Sa Internet digital word watermarking transmission having symmetrical algorithm authentication key before transmission and authentication key watermarking phase and transmission watermarked words
US7615234B2 (en) 2001-09-11 2009-11-10 Glide Pharmaceutical Technologies Limited Drug delivery technology
US20060122577A1 (en) 2001-09-26 2006-06-08 Poulsen Jens U Modular drug delivery system
SE0103211D0 (en) 2001-09-27 2001-09-27 Pharmacia Ab New formulations and use thereof
US6723077B2 (en) * 2001-09-28 2004-04-20 Hewlett-Packard Development Company, L.P. Cutaneous administration system
US6893655B2 (en) 2001-10-09 2005-05-17 3M Innovative Properties Co. Transdermal delivery devices
US20030087937A1 (en) 2001-10-15 2003-05-08 Nils-Olof Lindberg Nicotine and cocoa powder compositions
US20030119879A1 (en) 2001-10-15 2003-06-26 Thomas Landh Nicotine and chocolate compositions
US6723340B2 (en) 2001-10-25 2004-04-20 Depomed, Inc. Optimal polymer mixtures for gastric retentive tablets
TWI312285B (en) 2001-10-25 2009-07-21 Depomed Inc Methods of treatment using a gastric retained gabapentin dosage
US7429258B2 (en) 2001-10-26 2008-09-30 Massachusetts Institute Of Technology Microneedle transport device
US20040037879A1 (en) 2001-11-02 2004-02-26 Adusumilli Prasad S. Oral controlled release forms useful for reducing or preventing nicotine cravings
US8329217B2 (en) 2001-11-06 2012-12-11 Osmotica Kereskedelmi Es Szolgaltato Kft Dual controlled release dosage form
ES2284943T3 (en) 2001-11-16 2007-11-16 Medinnovation Ag MEDICINAL PUMPING DEVICE.
US7311693B2 (en) 2001-11-26 2007-12-25 Nilimedix Ltd. Drug delivery device and method
US20040052843A1 (en) 2001-12-24 2004-03-18 Lerner E. Itzhak Controlled release dosage forms
SE0104388D0 (en) 2001-12-27 2001-12-27 Pharmacia Ab New formulation and use and manufacture thereof
US6852741B2 (en) 2001-12-31 2005-02-08 University Of Florida Compositions and methods for treatment of neurological disorders
US20060167032A1 (en) 2002-01-16 2006-07-27 Galer Bradley S Pharmaceutical composition and method for treating disorders of the central nervous system
CA2473541A1 (en) * 2002-01-16 2003-07-31 Encapsulation Systems, Inc. Substance delivery device
US20030159702A1 (en) 2002-01-21 2003-08-28 Lindell Katarina E.A. Formulation and use manufacture thereof
US7004928B2 (en) 2002-02-08 2006-02-28 Rosedale Medical, Inc. Autonomous, ambulatory analyte monitor or drug delivery device
US20040033253A1 (en) 2002-02-19 2004-02-19 Ihor Shevchuk Acyl opioid antagonists
US6861066B2 (en) * 2002-03-11 2005-03-01 Health Plus International Inc. Method for the delivery of a biologically active agent
AU2003225735A1 (en) 2002-03-11 2003-09-29 Altea Therapeutics Corporation Transdermal drug delivery patch system, method of making same and method of using same
US20070168501A1 (en) 2002-03-29 2007-07-19 Cobb Nathan K Method and system for delivering behavior modification information over a network
US6780171B2 (en) 2002-04-02 2004-08-24 Becton, Dickinson And Company Intradermal delivery device
AR039336A1 (en) 2002-04-23 2005-02-16 Alza Corp TRANSDERMAL ANALGESIC SYSTEMS WITH REDUCED ABUSE POTENTIAL
WO2003092764A2 (en) 2002-04-29 2003-11-13 Kevin Marchitto Controlled release transdermal drug delivery
US7417013B2 (en) * 2002-05-01 2008-08-26 Mcneil-Ppc, Inc. Warming and nonirritating lubricant compositions and method of comparing irritation
EP1503744A1 (en) 2002-05-13 2005-02-09 Alexza Molecular Delivery Corporation Delivery of drug amines through an inhalation route
US20040166159A1 (en) 2002-05-29 2004-08-26 Chien-Hsuan Han Pharmaceutical dosage forms having immediate and controlled release properties that contain an aromatic amino acid decarboxylase inhibitor and levodopa
US7767698B2 (en) 2002-06-03 2010-08-03 Mcneil Ab Formulation and use thereof
AU2003251482A1 (en) 2002-06-10 2003-12-22 Euro-Celtique, S.A. Disposal systems of transdermal delivery devices to prevent misuse of the active agents contained therein
US8911781B2 (en) 2002-06-17 2014-12-16 Inventia Healthcare Private Limited Process of manufacture of novel drug delivery system: multilayer tablet composition of thiazolidinedione and biguanides
US8586079B2 (en) 2002-06-21 2013-11-19 Graphic Productions Inc. Promotional simulation for transdermal patch sampler
WO2004002571A1 (en) 2002-06-28 2004-01-08 Alza Corporation A reservoir for use in an electrotransport drug delivery device
US7196619B2 (en) 2002-06-29 2007-03-27 Neil Perlman Habit cessation aide
IL166687A0 (en) 2002-08-30 2006-01-15 Alza Corp Embossable and writable multilaminate backing construction
DE10240165A1 (en) 2002-08-30 2004-03-18 Disetronic Licensing Ag Dispensing unit for use in infusion pumps comprises reservoir for infusion liquid fitted with spring-loaded piston, toothed wheel cooperating with locking bar to prevent piston moving
US20040052851A1 (en) 2002-09-16 2004-03-18 Graff Allan H. Modified release oral dosage form
US20040259816A1 (en) 2002-10-01 2004-12-23 Pandol Stephen J. Compositions comprising plant-derived polyphenolic compounds and inhibitors of reactive oxygen species and methods of using thereof
US7399401B2 (en) 2002-10-09 2008-07-15 Abbott Diabetes Care, Inc. Methods for use in assessing a flow condition of a fluid
US8999372B2 (en) 2002-11-14 2015-04-07 Cure Pharmaceutical Corporation Methods for modulating dissolution, bioavailability, bioequivalence and drug delivery profile of thin film drug delivery systems, controlled-release thin film dosage formats, and methods for their manufacture and use
EP3473251B1 (en) 2002-12-20 2023-12-13 NicoNovum AB A nicotine-cellulose combination
US7887842B2 (en) 2003-02-07 2011-02-15 Teikoku Pharma Usa, Inc. Methods of administering a dermatological agent to a subject
AU2003900769A0 (en) 2003-02-21 2003-03-13 Francis Paul Galea Medication and smoking cessation watch
US20040253249A1 (en) 2003-04-02 2004-12-16 Rudnic Edward M. Pulsatile transdermally administered antigens and adjuvants
US7182955B2 (en) 2003-04-30 2007-02-27 3M Innovative Properties Company Abuse-resistant transdermal dosage form
SE0301320D0 (en) 2003-05-06 2003-05-06 Astrazeneca Ab Positive modulators of nicotinic acetylcholine receptors
US20050159419A1 (en) 2003-05-14 2005-07-21 Pharmacia Corporation Compositions of a cyclooxygenase-2 selective inhibitor and a central nervous system stimulant for the treatment of central nervous system damage
US8021334B2 (en) 2004-05-30 2011-09-20 Nilimedix Ltd. Drug delivery device and method
US7149574B2 (en) 2003-06-09 2006-12-12 Palo Alto Investors Treatment of conditions through electrical modulation of the autonomic nervous system
US20050048020A1 (en) 2003-07-07 2005-03-03 Wille John J. Novel topical delivery system for plant derived anti-irritants
US7231839B2 (en) * 2003-08-11 2007-06-19 The Board Of Trustees Of The Leland Stanford Junior University Electroosmotic micropumps with applications to fluid dispensing and field sampling
DE10338174A1 (en) 2003-08-20 2005-03-24 Lts Lohmann Therapie-Systeme Ag Transdermal drug formulations with drug combinations for the treatment of Parkinson's disease
US7282217B1 (en) 2003-08-29 2007-10-16 Kv Pharmaceutical Company Rapidly disintegrable tablets
WO2005023227A2 (en) 2003-09-08 2005-03-17 Pfizer Health Ab Nicotine formulations and use thereof
NZ546662A (en) 2003-10-20 2009-03-31 Teva Pharma Composition and dosage form for sustained effect of levodopa
EP1527792A1 (en) 2003-10-27 2005-05-04 Novo Nordisk A/S Medical injection device mountable to the skin
CA2544291C (en) * 2003-10-27 2013-01-08 University Of Basel Transdermal drug delivery method and system
EP1682061B1 (en) 2003-10-28 2017-03-15 Noven Pharmaceuticals, Inc. Transdermal drug delivery device
KR20070011240A (en) 2003-11-13 2007-01-24 알자 코포레이션 System and method for transdermal delivery
US8589174B2 (en) 2003-12-16 2013-11-19 Adventium Enterprises Activity monitoring
US20050266032A1 (en) 2003-12-17 2005-12-01 Sovereign Pharmaceuticals, Ltd. Dosage form containing multiple drugs
US20050141346A1 (en) 2003-12-30 2005-06-30 Rawls David K. Smoking cessation device
US20050151110A1 (en) * 2004-01-14 2005-07-14 Minor Barbara H. Fluoroether refrigerant compositions and uses thereof
US7867511B2 (en) 2004-01-23 2011-01-11 Travanti Pharma Inc. Abuse potential reduction in abusable substance dosage form
LT2351555T (en) 2004-02-23 2016-12-27 Euro-Celtique S.A. Abuse resistance opioid transdermal delivery device
IL160578A (en) 2004-02-25 2013-11-28 Newvaltech Knowledge Services And Invest Ltd Remote coaching service and server
CN1938004B (en) 2004-03-30 2011-12-21 欧洲凯尔特公司 Tamper resistant dosage form comprising an adsorbent and an adverse agent
US20050226921A1 (en) 2004-04-13 2005-10-13 Kortzebom Robert N Non-invasive analysis and controlled dosage transdermal active patch
DE102004019916A1 (en) 2004-04-21 2005-11-17 Grünenthal GmbH Anti-abuse drug-containing patch
DE102004022678A1 (en) 2004-05-05 2005-12-15 Humboldt-Universität Zu Berlin Method for controlling a volume flow of a liquid in an osmotic micropump and osmotic micropump
CN1332667C (en) 2004-05-26 2007-08-22 中国科学院生物物理研究所 Compound preparation for resisting Parkinson's disease
US7019622B2 (en) 2004-05-27 2006-03-28 Research In Motion Limited Handheld electronic device including vibrator having different vibration intensities and method for vibrating a handheld electronic device
EP1755733A4 (en) 2004-05-28 2010-04-21 Georgia Tech Res Inst Methods and devices for thermal treatment
US7537590B2 (en) 2004-07-30 2009-05-26 Microchips, Inc. Multi-reservoir device for transdermal drug delivery and sensing
EP1790375A4 (en) 2004-08-12 2009-06-17 Hisamitsu Pharmaceutical Co Transdermal drug administration apparatus with microneedle
US7780981B2 (en) 2004-09-13 2010-08-24 Chrono Therapeutics, Inc. Biosynchronous transdermal drug delivery
DE102004044578A1 (en) 2004-09-13 2006-03-30 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system with an adhesive layer, method for siliconizing a backing layer of the system and use of the backing layer
US8252321B2 (en) 2004-09-13 2012-08-28 Chrono Therapeutics, Inc. Biosynchronous transdermal drug delivery for longevity, anti-aging, fatigue management, obesity, weight loss, weight management, delivery of nutraceuticals, and the treatment of hyperglycemia, alzheimer's disease, sleep disorders, parkinson's disease, aids, epilepsy, attention deficit disorder, nicotine addiction, cancer, headache and pain control, asthma, angina, hypertension, depression, cold, flu and the like
MX2007004315A (en) 2004-10-08 2008-03-11 Noven Pharma Transdermal drug delivery device including an occlusive backing.
US8343538B2 (en) 2004-10-08 2013-01-01 Noven Pharmaceuticals, Inc. Compositions and methods for controlling the flux of a drug from a transdermal drug delivery systems
CN101160088B (en) 2004-10-29 2010-04-28 杨章民 Body temperature, respiration, heart sound, deglutition monitoring and medical treatment inquiry unit
US8999356B1 (en) 2004-12-03 2015-04-07 Omp, Inc. Drug delivery system
CN101080220A (en) 2004-12-17 2007-11-28 阿尔扎公司 Temperature modulation of transdermal drug delivery
US7625924B2 (en) 2004-12-22 2009-12-01 Memory Pharmaceuticals Corporation Nicotinic alpha-7 receptor ligands and preparation and uses thereof
CN101111281B (en) 2005-02-01 2013-02-06 因特利杰克特有限公司 Devices, systems, and methods for medicament delivery
US8226610B2 (en) 2005-02-01 2012-07-24 Intelliject, Inc. Medical injector with compliance tracking and monitoring
US8231573B2 (en) 2005-02-01 2012-07-31 Intelliject, Inc. Medicament delivery device having an electronic circuit system
US20060188859A1 (en) 2005-02-22 2006-08-24 Haim Yakobi Method and system for computer implemented personal counseling
US20070042026A1 (en) 2005-03-17 2007-02-22 Wille John J Prophylactic and therapeutic treatment of topical and transdermal drug-induced skin reactions
US8029927B2 (en) 2005-03-22 2011-10-04 Blue Spark Technologies, Inc. Thin printable electrochemical cell utilizing a “picture frame” and methods of making the same
US9233203B2 (en) 2005-05-06 2016-01-12 Medtronic Minimed, Inc. Medical needles for damping motion
US8722233B2 (en) 2005-05-06 2014-05-13 Blue Spark Technologies, Inc. RFID antenna-battery assembly and the method to make the same
US8688189B2 (en) 2005-05-17 2014-04-01 Adnan Shennib Programmable ECG sensor patch
WO2006127905A2 (en) 2005-05-24 2006-11-30 Chrono Therapeutics, Inc. Portable drug delivery device
WO2007013975A2 (en) 2005-07-20 2007-02-01 Pharmorx Inc. Composition containing an opioid agonist and a partial opioid agonist, preferably buprenorphine , for controlling abuse of medications
US9687186B2 (en) 2005-07-21 2017-06-27 Steadymed Ltd. Drug delivery device
US8566121B2 (en) 2005-08-29 2013-10-22 Narayanan Ramasubramanian Personalized medical adherence management system
US20070086275A1 (en) 2005-10-18 2007-04-19 Robinson Robert J Electronic reminder device
JP4355722B2 (en) 2005-11-17 2009-11-04 セイコーエプソン株式会社 Fluid transport device
US7988660B2 (en) 2005-12-20 2011-08-02 Eli Lilly And Company Needle-free injection device
US20070149952A1 (en) 2005-12-28 2007-06-28 Mike Bland Systems and methods for characterizing a patient's propensity for a neurological event and for communicating with a pharmacological agent dispenser
EP1815919A1 (en) * 2006-02-03 2007-08-08 Uponor Innovation Ab Making an elongated product
WO2008069972A2 (en) 2006-12-01 2008-06-12 Aradigm Corporation Systems for effecting cessation of tobacco use
US20180110768A1 (en) 2007-04-02 2018-04-26 Parkinson's Institute Methods and compositions for reduction of side effects of therapeutic treatments
EP1977746B8 (en) 2007-04-02 2014-09-24 Parkinson's Institute Methods and compositions for reduction of side effects of therapeutic treatments
WO2013006643A1 (en) 2011-07-06 2013-01-10 The Parkinson's Institute Compositions and methods for treatment of symptoms in parkinson's disease patients
US20140207047A1 (en) 2013-01-22 2014-07-24 Chrono Therapeutics, Inc. Transdermal drug delivery system and method
US10105487B2 (en) 2013-01-24 2018-10-23 Chrono Therapeutics Inc. Optimized bio-synchronous bioactive agent delivery system
US10213586B2 (en) 2015-01-28 2019-02-26 Chrono Therapeutics Inc. Drug delivery methods and systems
AU2016228779A1 (en) 2015-03-12 2017-09-07 Chrono Therapeutics Inc. Craving input and support system
JP2020500081A (en) 2016-09-28 2020-01-09 クロノ セラピューティクス インコーポレイテッドChrono Therapeutics Inc. Transdermal drug delivery device for delivering opioids
US20190374482A1 (en) 2016-12-05 2019-12-12 Chrono Therapeutics Inc. Transdermal drug delivery devices and methods
JP2020503950A (en) 2017-01-06 2020-02-06 クロノ セラピューティクス インコーポレイテッドChrono Therapeutics Inc. Device and method for transdermal drug delivery
WO2018148746A1 (en) 2017-02-13 2018-08-16 Chrono Therapuetics Inc. Transdermal drug delivery devices and methods
CA3101966A1 (en) 2018-05-29 2019-12-05 Morningside Venture Investments Limited Drug delivery methods and systems

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5538503A (en) * 1993-09-15 1996-07-23 Henley; Julian L. Programmable apparatus for reducing substance dependency in transdermal drug delivery

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009076363A1 (en) * 2007-12-10 2009-06-18 Isis Biopolymer Llc Iontophoretic drug delivery device and software application
DE102010030502A1 (en) 2010-06-24 2011-12-29 HSG-IMIT-Institut für Mikro- und Informationstechnologie Quellstoffaktor and conveyor, in particular for a medicinal active substance
WO2011161246A2 (en) 2010-06-24 2011-12-29 Hsg-Imit - Institut Für Mikro- Und Informationstechnologie Swellable substance actuator and delivery device, in particular for a medicinal active substance
WO2013136176A1 (en) * 2012-03-13 2013-09-19 Becton Dickinson France Injection device having a miniaturized drug delivery portion
US9844631B2 (en) 2012-03-13 2017-12-19 Becton Dickinson France Injection device having a miniaturized drug delivery portion
US10010676B2 (en) 2012-03-13 2018-07-03 Becton Dickinson France Method of manufacture for a miniaturized drug delivery device
US10080843B2 (en) 2012-03-13 2018-09-25 Becton Dickinson France Method of manufacture for a miniaturized drug delivery device
WO2017019526A2 (en) 2015-07-24 2017-02-02 Kimberly-Clark Worldwide, Inc. Methods for better delivery of active agents to tumors
EP3325080A4 (en) * 2015-07-24 2019-04-17 Sorrento Therapeutics, Inc. Methods for better delivery of active agents to tumors
US10806913B2 (en) 2015-07-24 2020-10-20 Sorrento Therapeutics, Inc. Methods for better delivery of active agents to tumors
EP3925599A1 (en) * 2015-07-24 2021-12-22 Sorrento Therapeutics, Inc. Methods for better delivery of active agents to tumors

Also Published As

Publication number Publication date
US20060062838A1 (en) 2006-03-23
US20100280432A1 (en) 2010-11-04
US20170100573A1 (en) 2017-04-13
CA2580329A1 (en) 2006-03-23
US9555227B2 (en) 2017-01-31
EP1802258A2 (en) 2007-07-04
AU2005284908B2 (en) 2011-12-08
JP5254616B2 (en) 2013-08-07
WO2006031856A3 (en) 2006-08-17
US20200330369A1 (en) 2020-10-22
US11471424B2 (en) 2022-10-18
EP1802258A4 (en) 2015-09-23
CA2580329C (en) 2015-01-06
US20140200525A1 (en) 2014-07-17
JP2008512215A (en) 2008-04-24
AU2005284908A1 (en) 2006-03-23
US20150283367A1 (en) 2015-10-08
US20210169822A1 (en) 2021-06-10
US7780981B2 (en) 2010-08-24
US20170100572A1 (en) 2017-04-13
US10716764B2 (en) 2020-07-21
US9555226B2 (en) 2017-01-31
US20150283366A1 (en) 2015-10-08
US20070191815A1 (en) 2007-08-16

Similar Documents

Publication Publication Date Title
AU2005284908B2 (en) Biosynchronous transdermal drug delivery
US10258778B2 (en) Biosynchronous transdermal drug delivery for longevity, anti-aging, fatigue management, obesity, weight loss, weight management, delivery of nutraceuticals, and the treatment of hyperglycemia, alzheimer's disease, sleep disorders, parkinson's disease, aids, epilepsy, attention deficit disorder, nicotine addiction, cancer, headache and pain control, asthma, angina, hypertension, depression, cold, flu and the like
US5415629A (en) Programmable apparatus for the transdermal delivery of drugs and method
AU2007238685B2 (en) Transdermal methods and systems for the delivery of anti-migraine compounds
WO2008054788A9 (en) Transdermal delivery techniques for drugs, nutraceuticals and other active substances
US20140207047A1 (en) Transdermal drug delivery system and method
AU2015203213B2 (en) Transdermal methods and systems for the delivery of anti-migraine compounds
AU2012261753A1 (en) Transdermal methods and systems for the delivery of anti-migraine compounds

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 BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK 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: 2007531458

Country of ref document: JP

Ref document number: 2580329

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005284908

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005796698

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005796698

Country of ref document: EP