WO2014207746A1 - Method and device for delivering therapeutic agents to a target region and means thereof - Google Patents

Method and device for delivering therapeutic agents to a target region and means thereof Download PDF

Info

Publication number
WO2014207746A1
WO2014207746A1 PCT/IL2014/050572 IL2014050572W WO2014207746A1 WO 2014207746 A1 WO2014207746 A1 WO 2014207746A1 IL 2014050572 W IL2014050572 W IL 2014050572W WO 2014207746 A1 WO2014207746 A1 WO 2014207746A1
Authority
WO
WIPO (PCT)
Prior art keywords
medication
drug
release
fluid
patient
Prior art date
Application number
PCT/IL2014/050572
Other languages
French (fr)
Inventor
Dan SADEH HOCHSTADTER
Yuval KAUFMAN
Original Assignee
Sadeh Hochstadter Dan
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 Sadeh Hochstadter Dan filed Critical Sadeh Hochstadter Dan
Publication of WO2014207746A1 publication Critical patent/WO2014207746A1/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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • 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
    • A61M25/00Catheters; Hollow probes
    • A61M25/10Balloon catheters
    • 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
    • A61M31/00Devices for introducing or retaining media, e.g. remedies, in cavities of the body
    • 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
    • A61M25/00Catheters; Hollow probes
    • A61M25/0043Catheters; Hollow probes characterised by structural features
    • A61M2025/0057Catheters delivering medicament other than through a conventional lumen, e.g. porous walls or hydrogel coatings
    • 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
    • A61M25/00Catheters; Hollow probes
    • A61M25/01Introducing, guiding, advancing, emplacing or holding catheters
    • A61M25/02Holding devices, e.g. on the body
    • A61M2025/0293Catheter, guide wire or the like with means for holding, centering, anchoring or frictionally engaging the device within an artificial lumen, e.g. tube
    • 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
    • A61M25/00Catheters; Hollow probes
    • A61M25/10Balloon catheters
    • A61M2025/1043Balloon catheters with special features or adapted for special applications
    • A61M2025/105Balloon catheters with special features or adapted for special applications having a balloon suitable for drug delivery, e.g. by using holes for delivery, drug coating or membranes
    • 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
    • A61M25/00Catheters; Hollow probes
    • A61M25/10Balloon catheters
    • A61M2025/1043Balloon catheters with special features or adapted for special applications
    • A61M2025/1086Balloon catheters with special features or adapted for special applications having a special balloon surface topography, e.g. pores, protuberances, spikes or grooves

Definitions

  • the present invention generally relates to a drug delivery device, system and method and more particu larly to biodegradable device tor delivering medications to at least one target region via the intraperitoneal (IP) route,
  • IP intraperitoneal
  • US patent No. 487 542 provides a method and a device tor delivering medicinal to animal and human bladders.
  • the device is a polymeric, miniceUular container surrounding an internal reservoir which contains the medicinal.
  • the device delivers the medicinal to the bladder at a prolonged, conti nuous and controlled rate.
  • EP patent application No. 0572932 discloses an implantable device that is capable of delivering a therapeutic agent to a tissue or organ over a long period ot time.
  • the implantable device is especially suited for treating tissues a nd orga ns that are comprised of smooth muscle.
  • the implantable device can deliver either a single therapeutic agent or a plurality of therapeutic agents to the tissue or organ at zero order kinetics.
  • US patent application No. 2010168563 discloses a device for application to a body cavity.
  • the device is insertable into the cavity of a patient.
  • the device includes a non-absorbable, flexible tube of an elongated shape, a removable core element situation within the tube, and a retention mechanism tor maintaining the device within the cavity,
  • US patent application No. 2006254580 describes a volume-adjustable device for the delivery of multiple medications to a patient, the device comprising multiple medication elements; an outer conta inment means having at least one open end into which the medication elements are removable inserted and a closure means removable and adjustably inserted into at least one end of the outer containment means is disclosed. It therefore remains a long felt and unmet need to provide novel means and methods for an device for delivering a drug in a more effective ana selective manner to the body cavity.
  • LDDDD implementable drug delivery device
  • the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the IP by the fluid.
  • DDD biocompatible drug delivery device
  • IP intraperitoneal
  • FS B l separation element having least one protuberance or/and indentation for distancing the hollow container from interior walls of the body cavity; further wherein the first and second chambers provide an ingress path for IP fluid to enter the hollow container and an exit path for expulsion of activated medication.
  • IP intraperitoneal
  • IP fluid contains an actuation element for activating the med ication; the actuation element is selected from the group consisting of IP fluid 's water, IP fluid 's electrolytes, IP fluid 's antibodies, IP flu id 's white blood cells, IP fluid 's bio-chemicals and combination thereof.
  • the drug-releasing component is a selective barrier element attached to at least first chamber selected from a group consisting of membrane, a mesh or a net like having at least one pore allowing some particles or chemicals to pass through.
  • the device as defined above ,wherein the device additionally comprising an anchoring means adapted for securing the device to a body tissue.
  • the medication is selected from a group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micron ized particle, particulate, pellet, core, powder, granu le, granulate, smal l mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
  • controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
  • biodegradable is selected from the group consisting of: liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chonaroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenyla lanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • a. providing biocompatible drug delivery device for activating and delivering at least one activated medication via the intraperitonea l (IP) route to at least one target region in a patient body comprising:
  • a hol low container having an externa l surface and interna l surface for housing an inactive medication, the inactive medication renderable active by contact with the IP fluid;
  • the method additionally comprising step of providing the device external surface with a functional separation element (FSE) having least one prot uberance or/and indentation for distancing the hollow container from interior walls of the body cavity the FSE; further wherein the method additionally comprising step of providing the chambers comprising an ingress path tor IP fluid to enter the hollow container and an exit path for expulsion of activated medication.
  • FSE functional separation element
  • IP intraperitoneal
  • controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
  • biodegradable is selected from the group consisting of: liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycolide (co-polymers of lactic acicl and glycolic acid) polyanhyclricles, polypeptides, hyaluronic acid, collagen, chonclroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • At least one drug-releasing component located between the chambers, adapted for controlling the release of the first and second medications from the chambers;
  • step of releasing of the medication via drug-releasing component further comprising step of associating the drug-releasing component with device's external surface and/or internal surface for controlling the rate of release of the medications; further wherein the method additionally comprising step of delivering the at least first medication and the at least second medication via the intraperitoneal (IP) route to at least one target region in a patient's body ; the at least one medication is actuated when in IP fluid connection.
  • IP intraperitoneal
  • the method additional ly comprising steps of delivering the at least one first medication to the intraperitoneal (IP) route, second to a first target region and third, to a second target region ; further wherein the step b comprising step of functionally associating the drug-releasing component with the device's external surface and/or internal surface for controlling the rate of release of the medications.
  • IP intraperitoneal
  • At least one first chamber and at least one second chamber for loading at least one first medication and at least one second medication respectively; and ii. at least one drug-releasing component located between the chambers adapted for med ication release from the chambers;
  • the method additionally comprising step of delivering the at least one first medication and the at least one second medication via the intraperitonea l (I P) route to at least two target regions in the patient's body; the at least one medication is actuated when in IP fluid connection.
  • I P intraperitonea l
  • the drug-releasing component is selected from the group consisting of: a selective barrier element attached to at least first chamber, selected from the group consisting of membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through.
  • the biodegradable from the group consisting of: liposomes, polylactides (poly!actic acid), polyg
  • the drug-releasing component is associated with the device external surface and/or internal surface for controlling the rate of release of the medications dosage; further wherein the at least one first medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body; the at least one medication is actuated when in IP fluid connection.
  • IP intraperitoneal
  • the drug-releasing component is a selective barrier element attached to at least first chamber, selected from the group consisting of membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through, but not other..
  • biodegradable is selected from the group consisting of: liposomes, polylactides, polyglycolide, poly (lactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • a biocompatible drug delivery device comprising:
  • At least one first chamber and at least one second chamber for loading the at least one first medication and the at least one second medication respectively;
  • an applicator adapted for affixing the device to the body tissue
  • the drug-releasing component is adapted for controlling the rate of release of the medication from device external surface or/and internal surface; further wherein the at least first medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body; the at least one medication is actuated when in I P fluid connection.
  • IP intraperitoneal
  • kits as defined in any of the above wherein the applicator is adapted for inserting the device into the body of a patient or/and for removing the device from said patient. It is another object of the present invention to provide the kit as defined in any of the above, wherein said device further comprising an anchoring means for securing said device to a body tissue.
  • DDP implementable drug delivery pump
  • At least one envelope with at least first layering surface for containing a medication said envelope comprises at ieast one ingress path for IP's fluid to both in let and outlet the layer for expulsion of the medication towards the IP by the fluid and further and
  • the medication delivery pump facilitates the deliver/ of the medication from the at least one envelope.
  • DDP implementable drug delivery pump
  • DDD implementable drug delivery device
  • IP intraperitoneal
  • the DDD comprising at least one envelope with at least first layering surface confining at ieast one first volume accomodating at least one first precursor and at least one second precursor; wherein the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid; and further wherein at least one first precursor and at least one second precursor forms an effective medication to target the region in a patient body.
  • DDD drug delivery device
  • FDDD feedback implementable drug delivery device
  • At least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication;
  • the envelope comprises at least one controllable path for IP's fluid to both inlet and outlet the layer for controlled expulsion of the medication towards the IP by the fluid;
  • controlling means regu lates the path thereby controlling the fluid flow.
  • FDDD feedback implementable drug delivery device
  • IP intraperitoneal
  • b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer; c. providing the least one first enveloping layer with an effective a releasing means, by selecting the releasing means from physical means, especially porosivicy parameters, and chemical means, especial ly solubility and selectivity;
  • d. providing, by the releasing means, the system with a drug's release profile, and e. selecting the profile from a group consisting of a parabolic release profi le, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer.
  • b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one or second enveloping layer.
  • b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one second drug with the at least one or second enveloping layer.
  • FIG. 1 presents the drug delivery device of the present invention
  • FIG, 2 presents a schematic view of the peritoneum surrounding the body organ
  • FIGs. 4A-B present a cross section view of the drug delivery device of the present invention
  • FIG. 5 presents a cross section view of the drug delivery device of the present invention
  • FIG. 6 presents a cross section view of the drug delivery device of the present invention
  • FIGs. 7-12 present a perspective view of a cluster-bomb-like implementable drug delivery systems of the present invention
  • FIGs. 13-19 present graphs of the release profile of the drug delivery device of the present invention
  • the present invention have been defined specifically to provide device, means ana method tor delivering a medication to a predefined location at the abdom inal cavity and more particularly to the peritoneal cavity of a patient for a complementary' medical analysis and treatment.
  • the present invention provides a device, kit and method for localized long-term administration of a therapeutic agent or a medication to a particu lar tissue or organ. T he device can be implanted or inserted once and release variety of drug dosages over an extended period.
  • the present invention provides an implanted device administrating a medication via the intraperitoneal (IP) route in patient's body cavity.
  • IP intraperitoneal
  • Using the IP rout for delivering a medication allows higher medication doses and more frequent administration of drugs, and it is more effective in killing ceils such as cancer cells in the peritoneal cavity, where ovarian cancer is li kely to spread or recur first.
  • the peritoneum is known as a membrane that surrounds the visceral organs forming the mesentery which connects the Bowel loops.
  • the physiologic characteristic of the peritoneal cavity provides a useful portal of entry in the body for variety pharmacological agents.
  • Several medications, such as antibiotics can be given via the intraperitoneal (IP) route to treat episodes of peritonitis .
  • IP intraperitoneal
  • the IP route can further be used for delivering a medication in patients with intra-abdominal malignancies, i.e. gynecological and gastrointestinal cancers.
  • the rate and amount of drug transfer in the peritoneum are dependent on several factors. Factors such as peritoneal inflammation, surface area, peritoneal blood flow, time of contact etc., influence the drug transfer.
  • IP route provides high concentration locally which targets the involved peritoneal tissues directly.
  • the peritoneum is known to support the abdominal organs and serves as a conduit for their blood and lymph vessels and nerves.
  • the structures in the abdomen are classified as intraperitoneal, retroperitoneal or intraperitoneal depending on whether they are covered with visceral peritoneum and whether they are attached by mesenteries (mensentery, mesocolon).
  • the IP fluid surrounds variety of organs such as the stomach, duodenum , jejunum, i leum, cecum, appendix, transverse colon, sigmoid colon, rectum, Liver, spleen, pancreas , uterus, fallopian tubes, ovaries , kidneys, adrenal glands, proximal ureters, renal vessels , urinary bladder and distal ureters, the IP can be an essential route for delivering medication to those organs.
  • organs such as the stomach, duodenum , jejunum, i leum, cecum, appendix, transverse colon, sigmoid colon, rectum, Liver, spleen, pancreas , uterus, fallopian tubes, ovaries , kidneys, adrenal glands, proximal ureters, renal vessels , urinary bladder and distal ureters
  • the IP can be an essential route for delivering medication to those organs.
  • the present invention further provides the usefulness of I P drug therapy device and the factors influencing it, as well as strategies to increase the efficacy, and conclude that IP route is an alternate route to the more conventional drug delivery routes, and can be successfu lly used when the target is within the peritoneal cavity or adjacent tissue.
  • the present invention further provides a device which after introducing into the peritoneal cavity, drug transfer occurs into the surrounding peritoneal tissues, and from there into the body compartments via the circulation.
  • Peritoneal transport occurs via three simultaneous processes: a) diffusion, b) ultrafiltration, and c) fluid reabsorption.
  • a three pore model has been validated, which suggests that transport across the peritoneal membrane occurs by pores of three different sizes.
  • the large pores and small pores with radius of about ⁇ 2 ⁇ responsible for the transport of larger and smaller sized solutes.
  • the absorption of intraperitoneailv admin istered macromolecules is linear in time, irrespective of molecular size or concentration.
  • the transport of the pharmacological agent across the peritoneum is governed by factors such as (a) dosing variables i.e. dose, volume, temperature, duration, composition of carrier solution etc., (b) drug properties i.e. molecular weight, io nic charge, apparent volume of d istribution (Vd), membrane binding, lipid/ water solubility etc, as well as (c) the characteristics of the peritoneum, i.e. surface area, charge, permeability, peritonitis, fibrosis, peritoneal blood flow and lymphatic absorption etc.
  • the rate and amount of drug transfer are dependent on several factors.
  • the kinetics of the drug, as well as factors such as peritoneal inflammation, surface area, peritoneal blood flow, time of contact, etc. influence the drug transfer.
  • Dedrick et ah, Pharmacokinetic Problems in Peritoneal Drug Administration-, Tissue Penetration and Surface Exposure JNCI ] Nad Cancer Inst (1997) 89(7): 480-487 described in a pharmacokinetic model which showed that with IP infusion the drug concentrations in the peritoneal cavity could theoretica lly exceed those in systemic circulation by three log values or more.
  • Maneuvers such as increasing the dwell duration, temperature or pressure, solutions of different ton icity 7 , use of vasoactive agents, and surfactants have been tried in an effort to increase the drug efficacy.
  • the surface area of the peritoneum which actually comes in contact with the therapeutic solution is a small fraction of the total peritoneal surface area.
  • newer icodextrin containing solutions can maintain the osmotic gradient for up to 24 hrs or longer, and have been used as a carrier for drugs such as 5FU for up to 96 hours.
  • Other factors such as the solution volume, patient size, and patient position, all affect the contact surface area.
  • An ambulator/ patient will have most of the fluid in the bottom of the cavity which decreases the contact area .
  • the device of the present invention has potential benefits for the use of IP route instead of using other routes such as intravenous (IV) route.
  • the IP route is with the ability to treat the patient on an outpatient basis, presence of an existing access for ad m inistration, avoidance of cost of IV lines, ana avoidance of IV route related toxicities such as phlebitis, and loss of veins which may need to be preserved.
  • I P route is an alternate manner to deliver a medication, especially where the target is contained within the peritoneal cavity. Gene therapy delivery may further use the IP route.
  • the device is configured to be insertable and/or implanted via natural body orifice or any opening to a cavity or passage of the body.
  • the drug is released into the surround ing of the peritoneal tissues and from there into the body compartments via the circulation. T he med ication rate and amount released are dependent on several factors such as the kinetics of the drug, peritoneal inflammation, surface area, peritoneal blood flow and time of contact, the medication adm inistrated vi the IP route provides high concentration locally of the medication which further targets the involved peritoneal tissues directly.
  • the major advantage of IP therapy is regional dose intensity. After intracavitary drug administration, the peritoneal cavity is exposed to higher concentrations compared to other parts of the body. The concentration differential occurs because drug movement from peritoneal cavity 7 to plasma (peritoneal clearance) is generally slow relative to drug clearance from the body.
  • the peritoneal cavity is the site of disease in several cancers, including ovarian, gastrointestinal and peritoneal mesothelioma.
  • Endometriosis a gynecological medical condition in which cells from the lining of the uterus (endometrium) appear and flourish outside the uterine cavity, most commonly on the membrane which lines the abdominal cavity.
  • the uterine cavity is lined with endometrial cells, which are under the influence of female hormones.
  • Endometrial-! ike cells in areas outside the uterus (endometriosis) are influenced by hormonal changes and respond in a way that is similar to the cells found inside the uterus. Symptoms often worsen with the menstrua l cycle.
  • Endometriosis is typically seen during the reproductive years. Symptoms may depend on the site of active endometriosis. Its main but not universal symptom is pelvic pain in various manifestations. Endometriosis is a common finding in women with infertility. Endometriosis can be treated in a variety of ways, includ ing pain medication, hormonal treatments, and surgery. In Endometriosis disease endometrial glands and stroma implant and grow in areas outside the uterus. The location a nd inflammatory response to these lesions are believed to play key role in symptoms ana signs associated with endometriosis.
  • the present invention provides an device which enables a medication delivery to a target region, organ or area related and affected of Endometriosis disease within the body cavity of a patient.
  • the device of the present invention may be further provided for the treatment of Endometriosis and for variety types of Cancer such as Ovarian, Peritoneal, Pseudomyxoma peritonei, Mucinous, adenocarcinoma of the appendix, Mesothelioma and Colorectal carcinoma.
  • the device may further be provided for treating variety of Infection such as: Peritonitis, P! L) - acute, PID - chronic and Tuberculosis.
  • the device may further be provided for treating Familial Mediterranean fever (FMF) ana for prevention of adhesions: Post-operative and Post-infectious.
  • FMF Familial Mediterranean fever
  • the device may further be provided for the treatment of extraperitoneal pathology: such as Inflammatory Bowel Disease- IB D (Crohn's disease and Ulcerative colitis) and reservoir for chronic system ic treatment.
  • controlled release system refers hereinafter to but is not limited to , drug delivery- systems (DDS), controlled release systems, delayed release systems, modified release systems, slow release systems, pH dependent systems, pH independent coatings systems, and any combinations thereof. It is further in the scope of the invention, wherein the term “controlled release” refers hereinafter to but is not limited to sustained release, sustained action, controlled release, extended action, timed release dosage forms and other parallel terms used to identify drug delivery systems that are designed to achieve a prolonged therapeutic effect by continuously releasing medication over an extended period of time after the administration of single dose.
  • Physically enabled systems are selected, in a non-limiting manner, from osmotic pressure- activated DDS; hydrodynamic pressure-activated DDS; vapor pressure-activated DDS; Mechanically activated DDS; magnetica lly activated DDS; mono-phoresis activated DDS; iontophoresis activated DDS; hydration-activated DDS etc.
  • Chemically enabled systems are selected, in a non-limiting manner, from pH- activated DDS; ion- activated DDS; hydrolysis- activated DDS etc.
  • Biochemically enabled systems are selected, in a non-limiting manner from enzyme- activated DDS and other biochemical- activated DDS.
  • an implemeritable drug delivery pump useful for facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body.
  • the DDP comprises a. at least one envelope with at least first layering surface for containing a medication; the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the IP by the fluid; and at least one first volume confined by the envelope, comprising effective measure of the med ication; and a medication delivery pump; wherein the medication delivery pump facilitates the delivery of the medication from the at least one envelope.
  • a method of facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body comprises a step of providing an implementable drug delivery pump (DDP) by containing a medication within at least one envelope with at least first layering surface; providing the envelope with at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the I P by the fluid; a step of confining by the envelope at least one first volume, comprising effective measure of the medication; and a step of facilitating the delivery of the medication from the at least one envelope by means of a medication delivery pump.
  • DDP implementable drug delivery pump
  • the DDS are in connection with or comprises drug's or fluid's pump such that a DDP is provided.
  • a DDP is provided.
  • an Alzet® - like osmotic pump is utilized.
  • Combitally available ALZET osmotic pumps and their like are miniature, infusion pumps for the continuous dosing.
  • another DDP is provided, namely a hydrodynamic pressure-activated drug delivery system (e.g., push- pull osmotic pump) is utilized.
  • This system is fabricated by enclosing a collapsible, impermeable container, which contains liquid drug formulation to form a drug reservoir compartment inside rigid shape-retaining housing.
  • a composite laminate of an adsorbent layer and a swellable, hyd roph ilic polymer layer is sandwiched.
  • another DDP is provided, namely a vapor pressure-activated drug delivery system is utilized.
  • the drug reservoir in a solution formulation is contained inside an infusate chamber. It is physically separated from the vapor pressure chamber by a freely movable bellows.
  • the vapor chamber contains a vaporizabie fluid, which vaporizes at body temperature and creates a vapor pressure. Under the vapor pressure created, the bellows moves upward & forces the drug solution in the infusate chamber to release, through a series of flow regulators and delivery cannula into the blood circulation at a constant flow rate.
  • DDP a mechanically activated drug delivery system
  • drug reservoir is in solution form retained in a container equipped with mechanically activated pumping system.
  • another DDP is provided, namely magnetically activated drug delivery system is utilized.
  • drug reservoir is a dispersion of peptide or protein powders in polymer matrix from, which macromolecular drug can be delivered only at a relatively slow rate. ' Ibis low rate of delivery can be improved by incorporating electromagneticaliy triggered vibration mechanism into polymeric device combined with a hemispherical design. Device is fabricated by positioning a tiny magnet ring in core of hemispherical drug dispersing polymer matrix.
  • This delivery device is fabricated by positioning a tiny magnet ring in core of hemispherical drug dispersing polymer matrix. T he external surface is coated with drug impermeable polymer (ethylene vinyl acetate or silicon elastomer) except one cavity at the center of the flat surface.
  • This delivery device used to deliver protein drugs such as bovine serum albumin, at a low basal rate, by a simple diffusion process under non triggering condition, As the magnet is activated to vibrate by external electromagnetic field, drug molecules are delivered at much higher rate.
  • another DDP is provided, namely sonophoresis - activated drug delivery system (phonophoresis) is utilized. This type of system utilizes ultrasonic energy to activate or trigger the delivery of drug from polymeric drug delivery device.
  • Non-degradable polymer ethylene vinyl acetate
  • bioerodiable polymer poly[bis(p-carboxyphenoxy) alkane an hydride.
  • iontophoresis activated drug delivery system is utilized. This type of system uses electrical current to activate and to modu late the diffusion of charged drug across biological membrane.
  • another DDP is provided, namely hydration activated drug delivery system is utilized, in this system, the drug reservoir is homogeneously dispersed in a swellable polymer matrix fabricated from a hydrophilic polymer (ethylene glyxomethacrylate). The release of drug is controlled by the rate of swelling of polymer matrix.
  • a pH- activated drug delivery system is utilized.
  • This type of chemically activated system permits targeting the delivery of drug only in the region with selected pH range. It fabricated by coating the drug-containing core with a pH - sensitive polymer combination. For instances, a gastric fluid labile drug is protected by encapsulating it inside a polymer membrane that resist the degradative action of gastric pH.
  • another DDP is provided, namely ion- activated drug delivery system is utilized. An ionic or a charged drug can be delivered by this method and this system are prepared by first compiexing an ionic drug with an ion-exchange resin containing a suitable counter ion.
  • a complex between a cationic drug with a resin having a SO 3 group or between an anionic drug with a resin having a N(CHi)3 group by forming a complex between a cationic drug with a resin having a SO 3 group or between an anionic drug with a resin having a N(CHi)3 group.
  • the granules of drug-resin complex are first treated with an impregnating agent & then coated with a water-insoluble but water-permeable polymeric membrane.
  • This membrane serves as a rate- controlling barrier to modulate the influx of ions as well as the release of drug from the system
  • an electrolyte medium such as gastric fluid ions diffuse into the system react with drug resin complex & trigger the release of ionic drug.
  • hydrolysis- activated drug delivery system is utilized. This type of system depends on the hydrolysis process to activate the release of drug.
  • Drug reservoir is either encapsulated in microcapsules or homogeneously dispersed in microspheres or nano particles tor injection. It can also be fabricated as an implantable device. All these systems can be prepared from bioerodib!e or biodegradable polymers (polyanhydride, polyorthoesters). It is activated by hydrolysis-induced degradation of polymer chain & is controlled by rate of polymer degradation.
  • DDP an enzyme - activated drug delivery system
  • This type of biochemical system depends on the enzymatic process to activate the release of drug.
  • Drug reservoir is either physically entrapped in microspheres or chemically bound to polymer chains from biopolymers (albumins or polypeptides).
  • the release of drug is activated by enzymatic hydrolysis of biopolymers (albumins or polypeptides) by specific enzyme in target tissue.
  • another DDP is provided, namely a feedback regulated drug delivery system is utilized. In this group the release of drug molecules from the delivery system is activated by a triggering agent. Rate of drug release is control led by concentration of triggering agent.
  • bioerosion-regulated drug delivery system namely bioerosion-regulated drug delivery system is utilized.
  • the system consisted of drug-dispersed bioerodible matrix fabricated from poly (vinyl methyl ether) ester which is coated with layer of immobilized urease. In a solution with near neutral pH, the polymer only erodes very slowly. In presence of urea, urease metabolizes urea to form ammonia. This causes increase in pH & rapid degradation of polymer with release of drug molecule.
  • bioresponsive drug deliver/ system Drug reservoir is contained in device enclosed by bioresponsive polymeric membrane whose drug permeability is controlled by concentration of biochemica l agent.
  • a lternatively or additional ly another DDP is provided, namely a self-regulating drug delivery system is utilized.
  • This type of system depends on a reversible & competitive binding mechanism to activate and to regulate the release of drug.
  • Drug reservoir is drug complex encapsulated within a semi permeable polymeric membrane. The release of drug from the delivery system is activated by the membrane permeation of biochemical agent from the tissue in which the system is located.
  • unit dosage form or “dose form” or “drug” or “medication” or “therapeutic agent” interchangeably refers hereinafter to a pharmaceutically acceptable forms of a liquid, gel, solid, gas or semisolid dosage forms drug selected from the group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates, Emulsion, Hydrogel, Molecular encapsulation, Soitgel, Solution, Suspension, Syrup, Tincture, Tisane or any combination thereof, in a sustained-release formulations.
  • the core of the unit dosage form may be coated with at least one functional coating or film which may modify the release properties of the formulation.
  • the unit dosage form of the present invention may further comprise d ifferent drug release profiles from d ifferent layers.
  • I ' he medications can be any compound that is biologically active and requires long term ad m inistration to a tissue or organ for maximum efficacy.
  • Therapeutic agents that can be used in accordance with the present invention include, but are not limited to, antimuscarinic agents, anticholinergic agents, antispasniodic agents, calcium, antagonist agents, potassium, channel openers, musculotropic relaxants, antineoplastic agents, polysynaptic inhibitors, and beta- ad renergic stimulators.
  • anticholinergic agents are propantheline bromide, imipramine, mepenzolate bromide, isopropamide iodide, clidinium bromide, anisotropine methyl bromide, scopolamine hyd rochloride, and their derivatives.
  • antimuscarinic agents include, but are not limited to, hyoscyamine sulfate, atropine, methantheline bromide, emepronium. bromide, anisotropine methyl bromide, and their derivatives.
  • polysynaptic inhibitors are baclofen and its derivatives.
  • 8-adrenergic stimulators are ter-butaline and its derivatives.
  • Examples of calcium antagonists are terodiline and its derivatives.
  • Examples of musculotropic relaxants include, but are not limited to, dicyclomine hydrochloride, flavoxate hydrochloride, papaverine hydrochloride, oxybutynin chloride, and their derivatives.
  • antineoplastic agents include, but are not limited to, carmusti ne levamisole hyd rochloride, flutamide, (w-methy!-N-[4-nitro-3-(tri.fluorom.ethy! phenyl]), adriarnycin, doxorubicin hydrochloride, idamycin, fluorouracil, Cytoxan, mutamycin, mustargen and leucovorin calcium.
  • the device may contain medication in a gas form (e.g Nitric Oxide,NO) such that when the inner structure of the device is in contact with the I P fluid a gas material is released.
  • a gas form e.g Nitric Oxide,NO
  • the gas material may further provide antimicrobial, anti- infection, and/or therapeutic activity and characteristics.
  • one of the uses for the control release delivery' of drugs to a specific tissue or organ is for internal organs that can be classified as mechanically dynamic organs.
  • mechanically dynamic organs include, but are not limited to, the stomach, intestines, heart, bladder, ureter, urethra, various sphincter muscles, and the esophagus.
  • the functions of these organs are maintained by their normal and timely mechanical contractions. Should the mechanical properties be lost, the intended functions of these organs are partially or completely lost- Reference is now made to Fig.
  • FIG. 1 which illustrates a biocompatible drug delivery device 1 for the delivery of at least a first med ication and at least second med ication to a patient body cavity, comprising: at least one first chamber 3 and at least one second chamber 4 for loading at least one first med ication and at least one second medication respectively, (b)at least one drug- releasing component 2 adapted for medication release; and optionally, (c)an anchoring means 6 adapted for securing the device to a body tissue.
  • the drug-releasing component is adapted for controlling the rate of release of the medication from the device internal surface or external surface.
  • the medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body.
  • IP intraperitoneal
  • the present invention further provides an implementable drug delivery device (DDL)) for delivering a medication via the intraperitoneal (I P) to a target region in a patient body, the DDD comprising:(a)at least one envelope with at least first layering surface for containing a medicamen; and, (b)at least one first volume confined by the envelope, comprising effective measure of the medication; wherein the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the I P by the fluid.
  • DDL implementable drug delivery device
  • the present invention further provides a drug delivery device (DDD) for activating and delivering at least one activated medication via the intraperitoneal (IP) route to a target region in a patient body
  • DDD drug delivery device
  • the DDD comprising: (a) a hollow container having an external surface and internal su rface for housing an inactive medication, the inactive med ication renderable active by contact with the IP fluid; (b) at least one first chamber for loading the medication into the hollow container; and, (c) at least one second chamber for releasing the at least one activated medication into the interior body cavity; wherein the external surface is provided with a functional separation element (FSE) having least one protuberance or/ and indentation for distancing the hollow container from interior walls of the body cavity; further wherein the first and second chambers provide an ingress path for IP fluid to enter the hollow conta iner and an exit path for expulsion of activated medication.
  • FSE functional separation element
  • the present invention further provides a biocompatible drug delivery device for the deliver/ of at least one medication to a target region within patient body cavity, comprising: (a) at least one first chamber and at least one second chamber for containing at least one first medication and at least one second medication respectively? (b) at least one drug-releasing component adapted for medication release, located between the chambers, and optiona lly, (c)an anchoring means adapted for securing the device to a body tissue.
  • the each of the chambers are easily connected or disconnected from the device when loading a medication especial ly in surgical or emergency procedure.
  • the med ication is delivered first to the intraperitoneal (IP) route second to a first target region and third to a second target region.
  • the drug-releasing component is adapted for controlling the rate of release of the medication when in IP fluid connection.
  • the drug transfer via the I P route achieves therapeutic efficacy in the region of interest while minimizing the systemic toxicities.
  • the present invention further provides an intraperitoneal (I P) administrable device useful for treating endometriosis, the device comprising: at least a first chamber and a second chamber for containing the at least a first medication and at least a second medication respectively; and , an anchoring means adapted for securing the device to a body tissue; wherein the device is configured for controlling the rate of release of the medication when the medication is delivered to a target region via the intraperitoneal (IP) route .
  • I P intraperitoneal
  • IP intraperitoneal
  • the drug-releasing component may be a selective barrier element (not shown) attached to at least first chamber containing at least one medication.
  • the separating element may be a membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through, but not other. T he selective barrier shape and structure may further control the time of release of each medication within each chamber.
  • the medication may further be based upon a controlled release delivery system of the medication.
  • the controlled release delivery system may be a slow release, immediate release, sustained release and combination thereof.
  • the drug-releasing component may further be a combination of a separating element and a controlled release delivery system of at least first medication.
  • the drug delivery device may be composed of a biodegradable material selected from the group consisting ot: liposomes, polylactides (polylactic acid), polyglycolide (polymer ot glycolic acid), poly (lactide co-glycolide (co-polymers ot lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chonclroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone container.
  • the drug delivery device contains at least first medication which is physically trapped, or covalently or ionically immobilized in a biodegrad able matrix.
  • the device is adapted for loading a medication having a dosage form selected from a group consisting ot aliquot, tablet, caplet, capsule, pi ll, bolus, particle, micronized particle, particulate, pellet, core, powder, granu le, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
  • the dosage form is further based upon controlled release delivery system.
  • the controlled release delivery ; system may be a slow release, immediate release, sustained release and combination thereof.
  • T herefore at least first medication is based upon a slow release manner whilst at least second medication is based upon immediate release manner.
  • at least first medication is based upon a slow release manner whilst at least second medication is based upon sustained release manner
  • the device is a 3D container having a cylinder shape, a tube like, a star like, a ball like, a roll like.
  • the device is composed of a semi permeable wall having at least one opening channel for IP fluid entrance and medication release.
  • the device is a non-absorbable, flexible container having an elongated shape.
  • the device is adapted to release at least first medication at a rate of at least approximately 0. 1 meg day whilst at least second medication is released at a rate of at least approximately 0.5 meg/day over a period of at least approximately one week.
  • the medications may be transported via a) diffusion, b) ultrafiltration, c) fluid re- absorption and combination thereof.
  • the device may contain medications for treating at least one diseases, disorder or condition having different dosages for long treatment.
  • the device is used for treating Endometriotic disease.
  • the device of the present invention is a volume-adjustable device. Thereby it may be adjusted to the volume or number or medication elements contained within the medication delivery device.
  • the medications for IP delivery for treating of endometriosis are selected in a non- limiting manner from the group consisting of Progestatives - e.g. dienogest, Progesterone antagon ists / modulators such as mifepristone, GN RH agon ists - e.g. leuprolide acetate, Aromatase inhibitors - e.g. letrezo!e, NTHEs - e.g. parecoxib, Anti-inflammatory', Anti- angiogenic DA - e.g. cabergoline, N F Kappa-b medications, Vitamin D, Estrogen and a combination or mixtures thereof.
  • the device may Last and provide treatment within patient's body cavity tor at least a week.
  • the device may comprise an anchoring means for maintaining the device with in the body cavity.
  • the device is flexible and shaped to be insertable and/or implanted into the body IP cavity.
  • the device may be preferably liquid impermeable or at least liquid sem i-permeable.
  • the device further comprises at least one opening through which the medication can be employed and/or release.
  • the medication is situated and released from at any one of the following locations: a. the external surface of the device, b. the internal surface of device within at least one of the chambers and, c. within a network of pores of wh ich the device may comprise.
  • the regions within the body cavity may be chosen from the group consisting of. rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra (including prostate), bladder and, intra-articuiar cavity.
  • the device is capable of deliver/ for local treatment and/or systemic treatment.
  • the device of the present invention may combine several medications in order to increase the therapy effect in several body regions or organs and further reduce side effects.
  • the device may contain variety of substance combinations, shapes or presentations (capsules, tablets, soft gels) without chemical interaction.
  • the device of the present invention provides controlled release of drug in vivo by diffusion of the drug out of a polymer and/or by degradation of the polymer over a predetermined period following administration to the patient.
  • the medication may be employed into the implanted device using a syringe or catheter.
  • the device may additionally comprise an applicator for affixing the device to the body tissue via anchoring means.
  • the applicator may further be used for reloading the device chambers with additional medications.
  • the applicator may further be used tor better implantation ot the device within the body cavity of the patient.
  • the applicator is configured to enable the device to collapse easily into the body cavity.
  • Fig 2 is a prior art, presenting the peritoneal cavity.
  • the peritoneum is a membrane that forms the lining of the abdom inal cavity or the coelom it covers most ot the intra-abdominal (or coelomic) organs in amniotes ana some invertebrates (annelids, for instance). It is composed of a layer of mesothelium supported by a thin layer of connective tissue.
  • the peritoneum both supports the abdominal organs ana serves as a conduit for their blood and lymph vessels and nerves.
  • the structures in the abdomen are classified as intraperitoneal, retroperitoneal or intraperitoneal depending on whether they are covered with visceral peritoneum and whether they are attached by mesenteries (mensentery, mesocolon).
  • Figs. 3a-c illustrate the configuration of a biocompatible drug delivery device for activating and delivering at least one activated medication via the intraperitoneal (IP) route to at least one target region in a patient body, comprising: (a)a hollow container having an external surface and internal surface for housing an inactive medication, the inactive med ication renderable active by contact with the IP fluid, (b) at least one first chamber tor loading the at least one inactive medication into the hollow container, and (c)at least one second chamber for releasing the at least one activated medication into the interior body cavity; wherein the external surface is provided with a functional separation element (FSE) having least one protruberance or/ and indentation for distancing the hollow container from interior walls ot the body cavity the FSE; further wherein the chambers provide an ingress path for IP fluid to enter the hollow container and an exit path for expulsion of activated medication.
  • FSE functional separation element
  • a biocompatible drug delivery device for the delivery of at least one first medication to patient's body cavity via the intraperitoneal (IP) route to at least one target region in a patient body, comprising: an implant for housing and releasing the medication, wherein the implant further comprises at least one chamber for containing the drug; the implant is adapted for introduction and placement into the Peritoneal cavity via the Douglas pouch, the belly-button or any other natural body enter. .
  • the interior walls referees to the device walls, the interior surface of the device or/ana the central surface of the device.
  • the release of the medication from the implant may be by an actuation element selected from a group consisting of: IP fluid water, IP fluid electrolytes, I P fluid antibodies, I P fluid white blood cells, IP fluid bio-chemicals or any of the martials which the IP fluid consist.
  • an actuation element selected from a group consisting of: IP fluid water, IP fluid electrolytes, I P fluid antibodies, I P fluid white blood cells, IP fluid bio-chemicals or any of the martials which the IP fluid consist.
  • the device may 7 further comprise a shielding material (not shown) as an external coating surrounding the device.
  • the shielding is adapted as a barrier or a septum to prevent contact of the device outer surface with the body tissue and furtherniore preventing increment of the concentration release of the medication.
  • ⁇ he shielding material may comprise plurality of holes.
  • ⁇ he device of the present invention may be inserted via the Douglas pouch, rectum, vagina or any other natural body enter.
  • the device of the present invention may 7 further be inserted placed and transported via other fluidly routes or humid regions such as the eye area, the digestive system, the bladder, the vagina or the like.
  • Fig 3c illustrates a cylindrical body configuration of the device of the present invention having an external surface comprising a plurality 7 of protrusion such as actuation flaps or pins allowing the movement of the device within the body cavity and further allowing the device transported with the IP fluid flow 7 to wonder around the organs.
  • the pins allow 7 movement of the device in the IP fluid and further to deliver at least one medication to a targeted region.
  • the device may comprise a monitor unit or a detecting unit for measuring physical or/and chemical characteristics or parameter with the device and fluid conditions such as temperature, humidity, pressure, pH, ionic change, osmolarity, concentration of substances such as acid-lactic, glucose, lipids, hormones, gases, enzymes, inflammatory mediators, plasmin, albumin, lactoferrin, creat.in.in, proteins and the like.
  • the monitor may be programed to begin or stop the release of the medication according to a pre-programed range of at least one of the mentioned parameters.
  • the monitor may further be programed to instruct and control the medication dosage release from the device.
  • the device may further comprise anchoring means for securing the device implanted to a wall of an organ within a patient body cavity.
  • the anchoring means may be located in the distal portion or the proximal portion of the device.
  • the illustrated configuration advantageously increases flexibility of the device and allow the device to be more effectively navigated through the IP fluid without damaging the device or injuring the patient tissues or organs.
  • Fig. 4a-h illustrates the inner surface of the biocompatible drug delivery device for the delivery of at least one first medication to patient's body cavity.
  • the device has an inner configuration comprising plurality of pores 42 providing a control release mechanism for the medication within the device.
  • the inner surface has a resilient structure perforated by pores through which the drug may be delivered from the chambers of the device.
  • the medicament may further be placed in any location or side within the device such that when the device is in fluid contact with the I P fluid the medication is released.
  • the device when placed in contact, with the patient's organ or I P fluid, provides active delivery of at least one medication into the IP fluid.
  • the pores may be expandable for enhancing the diffusion and the medication release from, the device.
  • the outer surface of the devise may comprise severa l layers having pores in different diameters in order to control the rate of the medication release from the device.
  • the device may comprise a first outer layer having pores with smal ler pore diameter and a second layer below the first layer having larger pore diameter for controlling the drug release from the device.
  • the drug may further be d ispersed in the polymer and re leasable when, the intra surface of the device is in contact with a liquid.
  • the rate of drug release may be dependent on the particular porous structure upon at least one outer layer of the device, which can be specifically formed to achieve a desired rate of release.
  • the pores may have round or other cross-sectional shapes and may have different, sizes.
  • a pore diameter refers to die average or ettective diameter of die cross- sections ot the pores. ⁇ he effective diameter of a cross-section that is not circular equals the diameter of a circular cross-section that has die same cross-sectional area as that of t e non-circular cross- section.
  • the pores may be filled with peritoneal fluid; the sizes of the pores may change dependi ng on the peritoneal fluid content in the device.
  • The. outer layer may thus behave like a sponge.
  • the pore diameter may be in the range from about 10 to 1 00 nm such that when the outer layer is in a dry condition wherein the peritoneal fl id content of the outer layer is at or near minimum .Furthermore, while a larger pore diameter may provide taster release initially, a smaller pore d iameter with increased lengt of interconnected pores can provide a more steady release at a similar rate.
  • the release rate may also be affected by the manner in which, the drug is incorporated in the device surface. When the drug is initially located in the device matrix, the rate may be slower. When the drug is initially located i the fluid, the rate may be higher.
  • the drug delivery rate may be controlled by controlling the pore structure and the manner in which the drug is incorporated in the device.
  • the diameter of the pores 41 may be between a 1 micrometer to about 1000 micrometers (1 mm), allowing exit of the medication in a dosage adjacent to the desired treatment.
  • Fig. 5 illustrates the cross section, of the external surface and the internal surface of the biocompatible drug delivery device for the delivery of at least one first medication to patient's body cavity.
  • the device may be composed of at least one medication or having different layer of several medications or different dosages of the same medication.
  • the internal surface of the device may be made of at least one medication 52 as an inner layer.
  • the device may further contain plurality of pores 52 allowing the entrance of the IP fluid and exit of the medication dissolved in the IP fluid.
  • Fig 5 further illustrates the outer layer of the device comprising plurality of pins adapted for preventing contact with the body tissue.
  • Fig 5 further illustrates the outer surface comprising plurality of protrusions having a pin like shape adapted to prevent any contact and further to distance the device from the patient's body tissue.
  • Fig, 6 presents a cross section view oi the biocompatible drug delivery device for the delivery of at least one medication to patient's body cavity.
  • the device comprises an outer shielding comprising plurality of pores 60 adapted to control the medication release from the device and in parallel preventing contact with the body tissue.
  • the device of Fig. 6 has a tubular configuration comprising an inner hollow tubu lar 61 structure allowing entrance of the IP fluid and an inner channel for loading at least one medication 62. T he device configuration allows the entrance of the I P fluid and exit of the medication dissolved in the IP fluid. The medication is released when in fluid contact with the IP fluid.
  • the device may be permeable or semipermeable to the IP fluid such that the IP fluid transports the med ication to a target region.
  • the device outer shielding prevents physical contact with the body tissue attached or anchored to the device and therefore preventing medication release with in the tissue.
  • the I P medication transport may occur via. pores having d ifferent sizes located in the outer shield ing of the device.
  • the absorption of intraperitoneally administered medication is linear in time, irrespective of molecular size or concentration.
  • the device of the present invention provides minimal fluctuations caused by temperature, humidity, pressure, pH, ionic change, osmolarity, concentration of su bstances such as acid-lactic, glucose, lipids, hormones, gases, en zymes, inflammatory mediators, plasmin, albumin, lactorerrin, creatinin, proteins and the like.
  • su bstances such as acid-lactic, glucose, lipids, hormones, gases, en zymes, inflammatory mediators, plasmin, albumin, lactorerrin, creatinin, proteins and the like.
  • the device is further Light weight and configured to prevent any friction with a surrounding tissue more particularly when the device is placed between organs.
  • the device is easily inserted and i n parallel easily retrieved.
  • the device is biocompatible and hypoallergenic therefore does not cause any side-effects.
  • the device is radiopaque therefore is opaque to one or another form of radiation, such as X-rays.
  • the device may further block radiation rather than allow it to pass through.
  • Table 1 below presents a variety of regions in the peritonea ! cavity which the device can be located in and further release a medication.
  • the present invention further provides a method for admi nostiring a dosage form to a patient, comprising steps of (a) providing a biocompatible drug delivery device for the delivery ot at least a first medication and at least second medication to a patient body cavity, comprising: (i)at least a first chamber and a second chamber for containing the at least one first medication and at least one second medication respectively, (iii) at least one drug-releasing component adapted for medication loading and release; optionally, (iv)an anchoring means adapted for securing the device to a body tissue; (b) inserting the drug delivery device to a body cavity of the patient, (c) optimally anchoring the drug delivery device to the body tissue of the patient; (d)releasing of the dosage form via drug-releasing component of the device into a the body cavity of the patient body.
  • the releasing of a dosage form via drug-releasing component is associated with the device internal surface or/and external surface for controlling the rate of release of the medication.
  • the method of the present invention further comprising the step of delivering the medication via the intraperitoneal (I P) route to at least two target regions in a patient body.
  • I P intraperitoneal
  • the present invention further provides a method of manufacturing a biocompatible drug delivery' device for the delivery of at least a first medication and a second medication to a patient body cavity, comprising steps or: (a) providing a container containing at least a first chamber and a second chamber for containing the at least a first medication and the second medication respectively; (b) attaching anchoring means to at least one edge of the container; (c) connecting at least one drug-releasing component to at least one first chamber, the drug- releasing component is adapted for medication loading and release, and (d) providing a drug- releasing component for medication release.
  • the drug-releasing component is associated with the device internal surface or/ and externa! surface for controlling the rate of release of the medication via the intraperitoneal (IP) route to at least two target regions in a patient abdominal cavity.
  • IP intraperitoneal
  • the device may be introduced via natura l body entrance, by injection procedure or by a surgical procedure.
  • the method as described above wherein the releasing the medication is from a first configuration suited for passage of the drug delivery device through the drug-releasing component to a second configuration suited for retaining the drug delivery device with the intraperitoneal (IP) direction route.
  • IP intraperitoneal
  • the medication dosage form is selected from a group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
  • the method as described above wherein the method comprising the step of providing a medication in a liquid phase, gas phase or solid phase.
  • the method as described above comprising the step of providing a medication based upon a controlled release delivery system.
  • controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
  • the device is a biodegradable container containing the medication that has been physically trapped, or covalently or ionically immobilized in the biodegradable matrix.
  • the biodegradable is selected from the group consisting of: liposomes, po!y!actides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycoltde (co-polymers of lactic acid and glycolic acid) po!yanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • the container is a cylinder or tube like implanted device.
  • the method as described above wherein the device is a non-absorbable, flexible container having an elongated shape.
  • the method as described above comprising the step of releasing the at least first medication at a rate of at least approximately 0.1 meg/day and at least second medication at a rate of at least 0,5 meg/day over a period of at least approximately one week.
  • the regions are selected from the group consisting of: rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity, eye and combination thereof.
  • the method as described above wherein the method comprising the step of transporting the medication to the Peritoneal fluid via a) diffusion, b) ultrafiltration, c) fluid re-absorption and combination thereof.
  • the present invention further provides a kit for the delivery of at least a first medication and a second medication to a patient body cavity, comprising: (a)a biocompatible drug delivery device comprising: (i) at least a first chamber and at least second chamber for loading and releasing the at least a first medication and the second medication respectively, (ii) optionally an anchoring means adapted for securing the device to a body tissue; and (iii) a drug-releasing component for controlling the rate of release of the medication, (b) an applicator adapted for affixing the device to the body tissue via anchoring means.
  • a biocompatible drug delivery device comprising: (i) at least a first chamber and at least second chamber for loading and releasing the at least a first medication and the second medication respectively, (ii) optionally an anchoring means adapted for securing the device to a body tissue; and (iii) a drug-releasing component for controlling the rate of release of the medication, (b) an applicator adapted for affixing the device
  • the device can travel and wander around the peritoneal cavity without any anchoring means.
  • At least first medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body.
  • IP intraperitoneal
  • the kit as described above, wherein the applicator is adapted for inserting the device into the body or an individual or/and for removing the device from the patient.
  • the kit as describee! above wherein the applicator is further adapted tor reloading the first medication and/or second medication into the device cham bers.
  • kits as described above, wherein the kit is useful for treating endometriosis.
  • an implementable drug delivery device useful for delivering a medication via the intraperitoneal (IP) to a target region in a patient body.
  • the DDD comprises at least one envelope with at least first layering surface confining at least one first volume accommodating at least one first precursor and at least one second precursor.
  • the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid.
  • At least one first precursor and at least one second precursor forms an effective medication to target the region in a patient body.
  • a method of targeting a region in a patient body comprises steps as follows: providing a drug delivery device (DDD) with at least one envelope, comprising at least first layering surface; confining within the envelope at least one first precursor and at least one second precursor; providing within the envelope at least one ingress path tor I P's fluid to both in let and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid; forming, by means of the at least one first precursor and at least one second precursor, an effective medication; and targeting the newly formed medication the region in a patient body.
  • DDD drug delivery device
  • the at least one first and at least one second precursors forms the medication(s) by reaction, polymerization, condensation, oxidation and reduction, complexation, acid-base reactions, precipitation, solid-state reactions, photochemical reactions, substitution, addition and elimination, rearrangement reaction, biochemical reactions etc.
  • the at least one first and at least one second precursors forms the medication(s) within the DDD, outside the DDD, adjacent the targeted organ, on the targeted organ etc.
  • FDDD feedbacked implementable drug delivery device
  • the FDD D comprises e.g., at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; the envelope comprises at least one controllable path for I P's f lu id to both inlet and outlet the layer for controlled expulsion of the medication towards the IP by the fluid; and a fluid's flow control ling means; wherein the controlling means regulates the path thereby controlling the fluid flow.
  • the fluid flow is selected from a group consisting of IP's fluid inflow, the medication's outflow or a combination of the same.
  • the device further comprises a sensing means in communication with the controlling means, for analyzing one or more mem bers of a group consisting of IP's parameters and targeted region of the body, thereby controlling the fluid flow in a feedbacked manner.
  • a method of delivering a time-resolved dose of one or more medications via. the intraperitoneal (IP) to a target region in a patient body comprises steps as follows: providing a feedbacked implementable drug delivery device (FDDD) with at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; the envelope comprises at least one controllable path for IP's fluid to both inlet and outlet the layer for controlled expulsion of the medication towards the IP by the fluid; further providing the FDDD with a fluid's f low controlling means; and regulating the path by means of the controlling means, thereby controlling the fluid flow.
  • FDDD feedbacked implementable drug delivery device
  • the method as defined above further comprises a step of selecting the fluid flow from a group consisting of IP's f lu id inf low, the medication's outf low or a combination of the same.
  • the method as defined above further comprises a step of communicating the sensing means with the controlling means, and one or more steps ol analyzing one or more members of a group consisting of IP's parameters and targeted region of the body, thereby controlling the fucid flow in a feedbacked manner.
  • the implementable drug delivery device as defined in any of the above, further comprising a pain relieving agents as medications, such as analgesic compositions.
  • a cluster-bomb-like implementable drug delivery system comprising at least one first envelope containing at least one first medication to be released from the envelope in predefined release profile; wherein the envelope is adapted to be deployed within the intraperitoneal (IP).
  • IP intraperitoneal
  • Figs 7 to 12 further illustrate a ciuster-bomb-like implementable drug delivery system having several envelopes to accommodate a at least one first medication .
  • the envelop may further adapted for loading and releasing at least one first medication and/or at least one second medication respectively.
  • at least one medication may be a controlled release medication.
  • the envelopes may be in a structure of a chamber, a cell, a shell, a carrier, or a matrix of medication.
  • the envelopes may contain at least one medication in each envelope or severa l medications (i.e., drugs) in one envelope.
  • the envelopes may be configured in a structure selected from a group consisting of a spherical shape (7a-b) , cylindrical shape (8a-b), a ring or tablet-like shape (9a-b, lOa-b) , a rectangu lar shape (8a- b), a tubular shape(lla-b-12a-b) a capsule shape, or any polygon known in the art.
  • the envelope may further comprise at least one aperture 100 for medications' release.
  • the envelope comprises a distal portion and a proximal portion whilst the proximal portion comprises at least one aperture adapted to be opened to permit medication to eject via the IP to a target region in a patient body.
  • the distal portion may comprise at least one aperture or may additiona lly comprise braking means adapted to bring the cluster bomb-like to a sudden deceleration or acceleration, whereby when the braking means bring the cluster bomb-like to the sudden deceleration or acceleration the at least one first medication is ejected through the proximal portion.
  • the device may further comprise at least one channel 110, 120 having a tube-like, ring-like or/and cylindrical shape for directing at least one medication to the evlepoe's aperture, thereby releasing the medication via the IP route.
  • the medication's release is characterized by a profile selected from the group consisting of: a parabo lic release profile, hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • the envelop is configured to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly- button, rectum, vagina and any combination thereof.
  • the envelop is configured to be introduced or implanted via a guiding tool such as catheter.
  • the profile is selected from the group consisting of a parabolic release profile, a hyperbolic release, profi le, an exponential release profile, a linear release profile and any combination thereof.
  • an envelope device for delivering at least one first control led release medication form the intraperitoneal (IP) to a target region in a patient body, the at least one first medication is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the envelop; wherein the controlled release is characterized by a profile selected from the group consisting of: a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body.
  • the system comprising at least one first enveloping layer and at least one first drug which is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the envelope; wherein the enveloping layer is characterized by a releasing means, and the releasing means are selected from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity; the releasing means provides the system with a drug's release profile selected from, a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • an implantable drug delivery system for delivering at least one first drug form the intraperitonea l (IP) to a target region located with a patient' body, as defined in any of the above is disclosed.
  • the system further eomp>rising at least one second enveloping layer in which the at least one first drug is contained, carried, doped, coated, d issolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the second enveloping layer.
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region, located with, a patient' body, as defined in any of the above is disclosed.
  • the system further comprising at least one second drug.
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed.
  • IP intraperitoneal
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed.
  • the system first drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an. exponential
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above.
  • IP intraperitoneal
  • the system is configured by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
  • the drug delivery system of claim 110 wherein the system is configured by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed.
  • the second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed.
  • IP intraperitoneal
  • the system first and second drug's release profile are different.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body comprising steps as follows: providing an implantable drug delivery system with at least one first enveloping layer and at least one first drug; containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer; providing the least one first enveloping layer with an effective a releasing means, by selecting the releasing means from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity; providing, by the releasing means, the system with a drug's release profile, and selecting the profile from a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed.
  • the method further comprising steps of providing the system with at least one second enveloping layer; and containing, carrying, eloping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is d isclosed .
  • the method further comprising step of providing the system at least one second drug.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed.
  • the method further comprising steps as follows: providing the system with at least one second enveloping layer; and containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one or second enveloping layer.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed.
  • the method further comprising steps as follows: providing the system with at least one second drug; and containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one second drug with the at least one or second enveloping layer.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed.
  • the method further comprising a step of providing the system's at least one first drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is d isclosed .
  • the method iurther comprising a step of providing the system's at least one first second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
  • a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed.
  • the method further comprising a step of configuring the drug delivery system by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
  • a method of implanting a drug delivery system as defined in any of the above id disclosed comprising step of introducing or implanting the same via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
  • a method of implanting a drug delivery system as defined in any of the above id disclosed The method step of configuring the system by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
  • a method of implanting a drug delivery system as defined in any of the above id disclosed further coniprising a step of configuring first and second drug's release profiles to be different.
  • Figs. 13 and 17 present a release polynomial profile thereby, the medication release rate (t) decreases for a period of time ,(ii) increases for a period of time , (iii) exhibits a zero order release for a period of time and a combination thereof.
  • the release profile may further include a parabolic release curve, a hyperbolic curve and a combination thereof, relative to time.
  • the release profile thereby, the release of the med ication may be adjusted by the medication controlled release system, or/and by the device configuration having apertures controlling the medication release rate relative to time.
  • Fig. 14 further presents a linear release profile thereby, the medication release rate may exhibits a zero order release for a period of time i.e constantly , increase linearly or decrease linearly with time.
  • Fig. 15 presents an exponential release profile thereby, the medication release rate decreases exponentially with time until the medication is exhausted.
  • Fig, 16 presents an exponential release profile thereby, the medication release rate increases exponentially with time as d e envelope and/or the medication moves forward to the target region.
  • Fig. 18-19 further illustrates a release profile of at least one first medication and a second medication or additional dosage based upon a controlled release system such that Fig. 18 presents an exponential release growth of one first med ication (red) and an exponential release decline (blue) of a second medication.
  • Fig. 19 presents a polynomial release profile of the first and second medication

Abstract

The present invention provides an implementable drug delivery device (DDL)) for delivering a medication via the intraperitoneal (IP) to a target region in a patient body and system and method thereof. The implementable drug delivery device comprising at least one envelope with at least first layering surface for containing a medication and at least one first volume confined by said envelope, comprising effective measure of the medication. The envelope further comprises at least one ingress path for I P's fluid to both inlet and outlet said layer for expulsion of said medication towards the IP by the fluid.

Description

METHOD AND DEVICE FOR DELIVERING THERAPEUTIC AGENTS TO A
TARGET REGION AND MEANS THEREOF
FIELD OF THE INVENTION
The present invention generally relates to a drug delivery device, system and method and more particu larly to biodegradable device tor delivering medications to at least one target region via the intraperitoneal (IP) route,
BACKGROUND OF THE INVENTION
Various modes for delivery of medicines to a mammalian body cavity are known in the art. Several patents describe delivery compositions and devices.
US patent No. 487 542 provides a method and a device tor delivering medicinal to animal and human bladders. The device is a polymeric, miniceUular container surrounding an internal reservoir which contains the medicinal. The device delivers the medicinal to the bladder at a prolonged, conti nuous and controlled rate.
EP patent application No. 0572932 discloses an implantable device that is capable of delivering a therapeutic agent to a tissue or organ over a long period ot time. The implantable device is especially suited for treating tissues a nd orga ns that are comprised of smooth muscle. The implantable device can deliver either a single therapeutic agent or a plurality of therapeutic agents to the tissue or organ at zero order kinetics.
US patent application No. 2010168563 discloses a device for application to a body cavity. The device is insertable into the cavity of a patient. The device includes a non-absorbable, flexible tube of an elongated shape, a removable core element situation within the tube, and a retention mechanism tor maintaining the device within the cavity,
US patent application No. 2006254580 describes a volume-adjustable device for the delivery of multiple medications to a patient, the device comprising multiple medication elements; an outer conta inment means having at least one open end into which the medication elements are removable inserted and a closure means removable and adjustably inserted into at least one end of the outer containment means is disclosed. It therefore remains a long felt and unmet need to provide novel means and methods for an device for delivering a drug in a more effective ana selective manner to the body cavity.
SUMMARY OF THE INVENTION
It is an object of the present invention to disclose an implementable drug delivery device (L)DD) for delivering a medication via the intraperitoneal (IP) to a target region in a patient body, the DDD comprising:
a. at least one envelope with at least first layering surface for containing a medication; and
b. at least one first volume confined by the envelope, comprising effective measure of the medication;
wherein the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the IP by the fluid.
It is an object of the present invention to disclose a biocompatible drug delivery device (DDD) for activating and delivering at least one activated medication via the intraperitoneal (IP) route to a target region in a patient body, the DDL) comprising:
a. a hollow container having an external surface and internal surface for housing an inactive medication, the inactive medication renderabie active by contact with the IP fluid;
b. at least one first chamber for loading the medication into the hollow container; and,
c. at least one second chamber for releasing the at least one activated medication into the interior body cavity;
wherein the external surface is provided with a functiona l separation element (FS B) having least one protuberance or/and indentation for distancing the hollow container from interior walls of the body cavity; further wherein the first and second chambers provide an ingress path for IP fluid to enter the hollow container and an exit path for expulsion of activated medication. It is an object of the present invention to disclose a biocompatible drug deliver/ device for the delivery of at least one first medication to patient's body cavity via the intraperitoneal (IP) route to at least one target region in a patient body, comprising: an implant for housing and releasing the medication, wherein the implant further comprises at least one chamber for containing the drug; the implant is adapted for introduction into the Peritoneal cavity.
It is another object of the present invention to provide the device as defined above, wherein the FSE is an outer coating for surrounding the device adapted as a shielding element; the outer coating is adapted for preventing contact of the device with the body tissue and further preventing increment in medication release.
It is another object of the present invention to provide the device as defined above, wherein the container has a 3 D shape selected from the group consisting of a tubular, a cylinder, a roll, a tube, a cube and combination thereof.
It is another object of the present invention to provide the device as defined above,wherein the ingress path and exit path are the same path or two separated paths.
It is another object of the present invention to provide the device as defined above wherein IP fluid contains an actuation element for activating the med ication; the actuation element is selected from the group consisting of IP fluid 's water, IP fluid 's electrolytes, IP fluid 's antibodies, IP flu id 's white blood cells, IP fluid 's bio-chemicals and combination thereof.
It is another object of the present invention to provide the device as defined above, wherein the external surface comprises at least one actuation flap movable by IP fluid flow; the flap allows movement of the device in the IP fluid and further promotes the medication delivery.
It is another object of the present invention to provide the device as defined above , wherein the drug-releasing component is a selective barrier element attached to at least first chamber selected from a group consisting of membrane, a mesh or a net like having at least one pore allowing some particles or chemicals to pass through.
It is another object of the present invention to provide the device as defined above ,wherein the device additionally comprising an anchoring means adapted for securing the device to a body tissue. It is another object of the present invention to provide the device as defined above,wherein the medication is selected from a group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micron ized particle, particulate, pellet, core, powder, granu le, granulate, smal l mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
It is another object of the present invention to provide the device as defined above, wherein the medication is in a liquid phase, gas phase, or solid phase.
It is another object of the present invention to provide the device as defined above, wherein additionally comprising a medication based upon a controlled release delivery system.
It is another object of the present invention to provide the device as defined above wherein the controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
It is another object of the present invention to provide the device as defined above, wherein the device is a biodegradable containing the medication that has been physically trapped, or covalently or ionicaliy immobilized in the biodegradable matrix.
It is another object of the present invention to provide the device as defined above wherein the biodegradable is selected from the group consisting of: liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chonaroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenyla lanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
It is another object of the present invention to provide the device as defined above, wherein the device is a nonabsorbable, flexible container having an elongated shape.
It is another object of the present invention to provide the device as defined above, wherein the at least first medication is at a rate of at least approximately 0. 1 meg/day over a period of at least approximately one week.
It is another object of the present invention to provide the device as defined above, wherein the device is used for treating Endometriotic disease. It is another object of the present invention to provide the device as defined above, wherein the device is semi permeable or permeable to the IP fluid.
It is another object of the present invention to provide the device as defined above , wherein the region is selected from the group consisting of: rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intta-articu lar cavity and combination thereof.
It is another object of the present invention to provide the device as defined above, wherein the medication is transported to the Peritoneal fluid via diffusion, ultrafiltration, fluid re- absorption and a combination thereof.
It is another object of the present invention to provide the device as defined above, wherein the device is affixed and secured to the body tissue using an applicator via anchoring means.
It is another object of the present invention to provide the device as defined above, wherein the medication is situated at any one of the fol lowing locations selected from the group consisting of: a. external surface of the device, b. internal surface of device within at least one of the chambers and, c. within a network of pores of which the device comprises.
It is another object of the present invention to provide the device as defined above, wherein further comprising a monitor unit for measuring physical or/ and chemical characteristics within the device; the monitor further adapted to control medication dosage release.
It is another object of the present invention to disclose a method for administering a medication to a patient, comprising steps of.
a. providing biocompatible drug delivery device for activating and delivering at least one activated medication via the intraperitonea l (IP) route to at least one target region in a patient body comprising:
i. a hol low container having an externa l surface and interna l surface for housing an inactive medication, the inactive medication renderable active by contact with the IP fluid;
ii. at least one first chamber for loading the at least one inactive medication into the hollow container; and, iii. at least one second chamber lor releasing the at least one activated medication into the interior body cavity;
b. introducing the drug delivery device to the patient's body cavity;
wherein the method additionally comprising step of providing the device external surface with a functional separation element (FSE) having least one prot uberance or/and indentation for distancing the hollow container from interior walls of the body cavity the FSE; further wherein the method additionally comprising step of providing the chambers comprising an ingress path tor IP fluid to enter the hollow container and an exit path for expulsion of activated medication.
It is another object of the present invention to provide the method as defined above, wherein additionally comprising step of providing the device having an outer coating surrounding the device as a shielding element; the outer coating is adapted for preventing contact with the body tissue and further preventing increment of the concentration release of the medication
It is another object of the present invention to provide the method as defined above, wherein the step pi providing the container having a 3 D shape selected from the group consisting of a tubular, a cylinder, a roil, a tube, a cube and a combination thereof.
It is another object of the present invention to provide the method as defined above, wherein additionally comprising step of providing at least one IP fluid's actuation element for activating the medication; the actuation element is selected from the group consisting of IP fluid 's water, IP fluid 's electrolytes, IP fluid 's antibodies, IP fluid's white blood cells, I P fluid 's bio-chemicals and combination thereof.
It is another object of the present invention to provide the method as defined above, wherein the method additional comprising step of providing the external surface having at least one protrusion or pin movable by IP fluid flow; the pin allow movement of the device in the IP fluid and further promoting the medication delivery.
It is another object of the present invention to provide the method as defined above, wherein the method additionally comprising step of anchoring and securing the drug delivery device to the body tissue of the patient using anchoring means It is another object of the present invention to provide the method as defined above, wherein the releasing the medication is from a first configuration suited for passage of the drug delivery device through the drug-releasing component to a second configuration suited for retaining the drug delivery device with the intraperitoneal (IP) direction route.
It is another object of the present invention to provide the method as defined above, wherein the medication is selected from a group consisting of aliquot, tablet, capiet, capsule, pill, bolus, particle, micronizecl particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
It is another object of the present invention to provide the method as defined above, wherein the method comprising the step of providing a medication in a liquid phase, gas phase or solid phase.
It is another object of the present invention to provide the method as defined above, wherein the method comprising the step of providing a medication based upon a controlled release delivery system.
It is another object of the present invention to provide the method as defined above, wherein the controlled release delivery system, is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
It is another object of the present invention to provide the method as defined above, wherein the device is a biodegradable container containing the medication that has been physically trapped, or covalently or ionically immobilized in the biodegradable matrix.
It is another object of the present invention to provide the method as defined above, wherein the biodegradable is selected from the group consisting of: liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycolide (co-polymers of lactic acicl and glycolic acid) polyanhyclricles, polypeptides, hyaluronic acid, collagen, chonclroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone. It is another object of the present invention to provide the method as defined above, wherein the method additionally comprising steps of releasing the at least first medication at a rate of at least approximately 0.1 meg/day and at least second medication at a rate of at ieast 0.5 meg,/ day over a period of at least approximately three months.
It is another object of the present invention to provide the method as defined above, wherein the the method additional ly comprising steps of providing the device for treating Endometriotic disease.
It is another object of the present invention to provide the method as defined above, wherein the the method additionally comprising steps of providing the device having a semi permeable characteristics.
It is another object of the present invention to provide the method as defined above, wherein the method additionally comprising steps of selecting the region from the group consisting of: rectum, large intestine, smaiiintestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear cana l, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity and combination thereof.
It is another object of the present i nvention to provide the method as defined above, wherein the method additionally comprising the step of transporting the medication to the Peritoneal fluid via a) diffusion, b) ultrafiltration, c) fluid re-absorption ana combination thereof.
It is another object of the present invention to provide the method as defined above, wherein additionally comprising step of affixing and securing the device to the body tissue using an applicator via anchoring means.
It is another object of the present invention to provide the method as defined in any of the above, wherein further comprising step of providing a monitor unit within the device for measuring physical or/and chemica l characteristics within the device; the monitor further adapted to control medication dosage release.
It is another object of the present invention to disclose a method method for administering a medication to a patient, comprising steps of: a. providing a biocompatible drug delivery device tor the delivery of at least one first medication and at least one second medication to a patient's body cavity, the device comprising:
i. at least first chamber and at least one second chamber for loading at least one first medication and the at least one second medication respectively; and,
ii. at least one drug-releasing component located between the chambers, adapted for controlling the release of the first and second medications from the chambers;
b. inserting the drug delivery device to a body cavity of the patient; and
c. releasing of the medication via the drug-releasing component into the body cavity of the patient's body;
wherein the step of releasing of the medication via drug-releasing component further comprising step of associating the drug-releasing component with device's external surface and/or internal surface for controlling the rate of release of the medications; further wherein the method additionally comprising step of delivering the at least first medication and the at least second medication via the intraperitoneal (IP) route to at least one target region in a patient's body ; the at least one medication is actuated when in IP fluid connection. is another object of the present invention to d isclose a method for administering a edication to a target region within patient's body cavity, comprising steps of:
a. providing a biocompatible drug delivery device for the delivery of at least a first medication ana a second medication to a patient body cavity, the device comprising:
i. at least one first chamber and at least one second chamber for loading at least one first medication and at least one second medication respectively; ana,
ii. at least one drug-releasing component located between the chambers adapted for medication release from the chambers;
b. inserting the d rug delivery device to a body cavity of the patient; and
c. releasing of the medication via drug-releasing component of the device into the body cavity of the patient body; wherein the method additional ly comprising steps of delivering the at least one first medication to the intraperitoneal (IP) route, second to a first target region and third, to a second target region ; further wherein the step b comprising step of functionally associating the drug-releasing component with the device's external surface and/or internal surface for controlling the rate of release of the medications.
It is another object of the present invention to disclose a method of manufacturing a biocompatible drug delivery device for the delivery of at least one first med ication and at least one second medication to a patient's body cavity, comprising steps of:
a. providing a biocompatible drug delivery device for the delivery of at least a first medication and a second medication to a patient body cavity, the device comprising:
i. at least one first chamber and at least one second chamber for loading at least one first medication and at least one second medication respectively; and ii. at least one drug-releasing component located between the chambers adapted for med ication release from the chambers;
b. connecting at least one drug-releasing component between the at least one first chamber and the at least one second chamber ; the drug-releasing component is for medication release;
wherein additionally comprising steps of associating the drug-releasing component with the at least device external surface and/or internal surface for controlling the rate of release of the at least first medication via the intraperitoneal (IP) route to at least two target regions in a patient's abdominal cavity.
It is another object of the present invention to disclose a method for administering a device to a target region within a patient's body cavity, comprising steps of:
a. providing a biocompatib le drug delivery device for the delivery of at least one first medication and at least one second medication to the intraperitoneal (IP) route, the device comprising:
i. at least one first chamber and at least one second chamber for loading at least one first medication and at least one second medi cation_respectively; and,
ii. at least one drug-releasing component located between the chambers adapted for medication release from the chambers; and, b. providing an applicator adapted for loading the device ;
c. placing the drug delivery device in the end of the applicator ;
d. inserting the end of the applicator to a target region within patient's body cavity; e. releasing the device from the end of the applicator whereby the device automatical ly seats itself within the patient's body cavity; and,
f. withdrawing the applicator from the patient's body cavity;
wherein the method additionally comprising step of delivering the at least one first medication and the at least one second medication via the intraperitonea l (I P) route to at least two target regions in the patient's body; the at least one medication is actuated when in IP fluid connection.
It is another object of the present invention to provide the method as defined in any of the above, wherein the drug-releasing component is selected from the group consisting of: a selective barrier element attached to at least first chamber, selected from the group consisting of membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additionally comprising step of providing high concentration of the medication when located in the IP route.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additionally comprising step of anchoring the drug delivery device to the body tissue of the patient using anchoring means.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additionally comprising step of attaching anchoring means to at least one edge of the device.
It is another object of the present invention to provide the method as defined in any of the above, wherein the step of releasing the medication is from a first configuration suited for passage of the drug delivery device through the drug-releasing component to a second configuration suited for retaining the drug delivery device with the intraperitoneal (IP) direction route. It is another object of the present invention to provide the method as defined in any of the above, wherein the the method additionally comprising steps of selecting the medication from a group consisting of aliquot, tablet, capiet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additionally comprising step of providing a medication in a liquid phase, gas phase or solid phase.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additionally comprising step of providing a medication based upon a controlled release delivery system.
It is another object of the present invention to provide the method as defined in any of the above, wherein the additionally comprising step of selecting the controlled release delivery system from the group consisting of: slow release, immediate release, sustained release and combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, wherein the step of providing the device having a biodegradable container containing the medication that has been physically trapped, or covalently or ionically immobilized in the biodegradable matrix.
It is another object of the present invention to provide the method as defined in any of the above, wherein the the method additionally comprising steps of selecting the biodegradable from the group consisting of: liposomes, polylactides (poly!actic acid), polyg!yco!ide (polymer of glycolic acicl), poly (lactide co-giycolide (co-polymers of lactic acid and glycoiic acid) poiyan hydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acicls, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone silicone and any combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, wherein the step of providing the device comprising a container having a 3 D shape selected from the group consisting of a tubu lar, a cylinder, a roll, a tube, a cube and combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, wherein the step of providing the device as a non-absorbable, flexible container having an elongated shape.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additionally comprising step of releasing the at least first medication at a rate of at least approximately 0.1 meg/day and at least second medication at a rate of at least 0.5 meg/day over a period of at least approximately one week.
It is another object of the present invention to provide the method as defined in any of the above, wherein the device is used for treating Endometriotic disease.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method additional ly comprising steps of selecting the region from the group consisting of: rectum, large intestine, smallintestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity and combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method comprising the step of transporting the medication to the Peritoneal fluid via a) diffusion, b) ultrafi ltration, c) fluid re-absorption and combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, wherein additionally comprising step of affixing and securing the device to the body tissue using an applicator via anchoring means.
It is another object of the present invention to disclose a biocompatible drug delivery device for the deliver/ of at least one first medication and at least one second medication to patient's body cavity, comprising: a. at least one first chamber and at least one second chamber for loading the at least one first medication and the at least one second medication respectively; and, b. at least one drug-releasing component located between the chambers adapted for medication release from the chambers;
wherein the drug-releasing component is associated with the device external surface and/or internal surface for controlling the rate of release of the medications dosage; further wherein the at least one first medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body; the at least one medication is actuated when in IP fluid connection.
It is another object of the present invention to provide the device as defined in any of the above, wherein the drug-releasing component is a selective barrier element attached to at least first chamber, selected from the group consisting of membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through, but not other..
It is another object of the present invention to provide the device as defined in any of the above, wherein the device is biodegradable .
It is another object of the present invention to provide the device as defined in any of the above, wherein further comprising an anchoring means lor securing the device to a body tissue.
It is another object of the present invention to provide the device as defined in any of the above, wherein the medication is selected from a group consisting of aliquot, tablet, caplet, capsu le, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates and any combination thereof
It is another object of the present invention to provide the device as defined in any of the above, wherein the medication is selected from a group consisting of aliquot, tablet, caplet, capsule, pi ll, bolus, particle, micronized particle, particulate, pellet, core, powder, granu le, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates and any combination thereof.
It is another object of the present invention to provide the device as defined in any of the above, wherein the medication is based upon controlled release delivery system. It is another object of the present invention to provide the device as defined in any of the above, wherein the controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device is a biodegradable container containing the medication that has been physically trapped, covalently or ionically immobilized in a biodegradable matrix.
It is another object of the present invention to provide the device as defined in any of the above, wherein the biodegradable is selected from the group consisting of: liposomes, polylactides, polyglycolide, poly (lactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
It is another object of the present invention to provide the device as defined in any of the above, wherein the container is a 3D shape selected from the group consisting of a tubular, a cylinder, a roll, a tube, a cube and combination thereof.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device is a non-absorbable, flexible container having an elongated shape.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device is semi permeable.
It is another object of the present invention to provide the device as defined in any of the above, wherein the at least first medication is released at a rate of at least approximately 0.1 meg,/ day whilst the at least second medication is released at a rate of at least approximately 0.5 meg/day over a period of at least approximately one week.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device is used for treating Endometriotic disease.
It is another object of the present invention to provide the device as defined in any of the above, wherein the regions are selected from the group consisting of: rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fa llopian tubes, urethra, bladder and, intra-articular cavity and combination thereof.
It is another object of the present invention to provide the device as defined in any of the above, wherein the at least first the medication is transported via a) diffusion, b) ultrafiltration, c) fluid re-absorption and combination thereof.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device is useful for treating endometriosis
It is another object of the present invention to provide the device as defined in any of the above, wherein the device additional ly comprising an applicator for affixing the device to the body tissue via anchoring means.
It is another object of the present invention to disclose a kit for the delivery of at least a first medication and a second medication to a patient body cavity?, comprising:
a. a biocompatible drug delivery device comprising:
i. at least one first chamber and at least one second chamber for loading the at least one first medication and the at least one second medication respectively;
ii. at least one drug-releasing component located between the chambers adapted for medication release from the chambers; and,
b. an applicator adapted for affixing the device to the body tissue;
wherein the drug-releasing component is adapted for controlling the rate of release of the medication from device external surface or/and internal surface; further wherein the at least first medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body; the at least one medication is actuated when in I P fluid connection.
It is another object of the present invention to provide the kit as defined in any of the above, wherein the applicator is adapted for inserting the device into the body of a patient or/and for removing the device from said patient. It is another object of the present invention to provide the kit as defined in any of the above, wherein said device further comprising an anchoring means for securing said device to a body tissue.
It is another object of the present invention to provide the kit as defined in any of the above, wherein said applicator is further adapted tor reloading said first medication and/or second medication into said device chambers.
It is another object of the present invention to provide the kit as defined in any of the above, thewherein said kit is useful tor treating endometriosis.
It is another object of the present invention to disclose an implementable drug delivery pump (DDP) for facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body, the DDP comprising:
a. at least one envelope with at least first layering surface for containing a medication; said envelope comprises at ieast one ingress path for IP's fluid to both in let and outlet the layer for expulsion of the medication towards the IP by the fluid and further and
b. at least one first volume confined by the envelope, comprising effective measure of the medication; and
c. a medication delivery pump;
wherein the medication delivery pump facilitates the deliver/ of the medication from the at least one envelope.
It is another object of the present invention to disclose a method of facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body, the method comprising: a. providing an implementable drug delivery pump (DDP) by contain ing a medication within at ieast one envelope with at least first layering surface; providing the envelope with at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the IP by the fluid;
b. confining by the envelope at least one first volume, comprising effective measure of the medication; and c. facilitating the delivery of the medication from the at ieast one envelope by means of a medication delivery pump.
It is another object of the present invention to disclose an implementable drug delivery device (DDD) for delivering a medication via the intraperitoneal (IP) to a target region in a patient body, the DDD comprising at least one envelope with at least first layering surface confining at ieast one first volume accomodating at least one first precursor and at least one second precursor; wherein the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid; and further wherein at least one first precursor and at least one second precursor forms an effective medication to target the region in a patient body.
It is another object of the present invention to disclose a method of targeting a region in a patient body, comprising:
a. providing a drug delivery device (DDD) with at least one envelope, comprising at least first layering surface;
b. confining within the envelope at least one first precursor and at least one second precursor;
c. providing within the envelope at least one ingress path tor IP's fluid to both inlet and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid;
d. forming, by means of the at least one first precursor and at least one second precursor, an effective medication; and
e. targeting the newly formed medication the region in a patient body.
It is another object of the present invention to disclose a feedback implementable drug delivery device (FDDD) for delivering a time-resolved dose of one or more medications via the intraperitoneal (IP) to a target region in a patient body, the FDDD comprising:
d. at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; the envelope comprises at least one controllable path for IP's fluid to both inlet and outlet the layer for controlled expulsion of the medication towards the IP by the fluid;
e. a fluid's flow controlling means; wherein the controlling means regu lates the path thereby controlling the fluid flow.
It is another object of the present invention to provide the FD DD as defined in any of the above, wherein the fluid flow is selected from a group consisting of IP's fluid inflow, the medication's outflow or a combination of the same.
It is another object of the present invention to provide the FD DD as defined in any of the above, wherein the device further comprising a sensing means in communication with the controlling means, for analyzing one or more members of a group consisting of IP's parameters and targeted region of the body, thereby controlling the fluid flow in a feedback manner.
It is another object of the present invention to disclose a method of delivering a time-resolved dose of one or more medications via the intraperitoneal (IP) to a target region in a patient body, the method comprising:
a. providing a feedback implementable drug delivery device (FDDD) with at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; the envelope comprises at least one controllable path for IP's flu id to both in let and outlet the layer for controlled expulsion of the medication towards the IP by the fluid;
b. further providing the FDDD with a fluid's flow controlling means; and
c. regulating the path by means of the controlling means, thereby controlling the fluid flow.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method further comprising a step of selecting the fluid flow from a group consisting of IP's fluid inflow, the medication's outflow or a combination of the same.
It is another object of the present invention to provide the method as defined in any of the above, wherein the method further comprising a step of communicating the sensing means with the controlling means, and one or more steps of analyzing one or more members of a group consisting of IP's parameters and targeted region of the body, thereby controlling the fluid flow in a feedback manner. It is another object of the present invention to provide the device as defined in any of the above, wherein the device further comprising radiopaque materials, especially metals, barium sulfate or iodine.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device further comprising a net-like layer facilitating the controlled release of the medication.
It is another object of the present invention to provide the device as defined in any of the above, wherein the device further comprising a net-like layer facilitating the controlled release of the medication.
It is another object of the present invention to provide the device as defined in any of the above, wherein medication is pain relieving agent, such as analgesic compositions.
It is another object of the present invention to disclose a implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, comprising at least one first enveloping layer and at least one first drug which is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the envelope; wherein the enveloping layer is characterized by a releasing means, and the releasing means are selected from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity; the releasing means provides the system with a drug's release profi le selected from a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein the system further comprising at least one second enveloping layer in which the at least one first drug is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the second enveloping layer.
It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein the system further comprising at least one second drug. It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein at least one of the second drugs is contained, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with in the first or second enveloping layers,
It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein the first drug's release prof le is selected from a group consisting ot a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein the system is configured by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
It is another object of the present invention to provide the drug delivery system as defined in any or the above, wherein the system is configured by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein the second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
It is another object of the present invention to provide the drug delivery system as defined in any of the above, wherein the first and second drug's release profile are different.
It is another object of the present invention to disclose a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient's body, comprising: a. providing an implantable drug delivery system with at least one first enveloping layer and at least one first drug;
b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer; c. providing the least one first enveloping layer with an effective a releasing means, by selecting the releasing means from physical means, especially porosivicy parameters, and chemical means, especial ly solubility and selectivity;
d. providing, by the releasing means, the system with a drug's release profile, and e. selecting the profile from a group consisting of a parabolic release profi le, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, further comprising:
a. providing the system with at least one second enveloping layer.and
b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer.
It is another object of the present invention to provide the method as defined in any of the above, further comprising step of providing the system at least one second drug.
It is another object of the present invention to provide the method as defined in any of the above, further comprising steps as follows:
a. providing the system with at least one second enveloping layer; and
b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one or second enveloping layer.
It is another object of the present invention to provide the method as defined in any of the above, further comprising steps as follows:
a. providing the system with at least one second drug; and
b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one second drug with the at least one or second enveloping layer.
It is another object of the present invention to provide the method as defined in any of the above, further comprising a step of providing the system's at least one first drug's release profile is selected from a group consisting ot a parabolic, release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof. It is another object of the present invention to provide the method as defined in any of the above, further comprising a step of providing the system's at least one first: second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
It is another object of the present invention to provide the method as defined in any of the above, further comprising a step of configuring the drug delivery system by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
It is another object of the present invention to disclose a method of implanting a drug delivery system as defined in claim 110 comprising step of introducing or implanting the same via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
It is another object of the present invention to disclose a method of implanting a drug delivery system as defined in claim 1 10 comprising step of configuring the system by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
It is another object of the present invention to provide the method as defined in any of the above, further comprising a step of configuring first and second drug's release profiles to be d ifferent.
BRIEF DESCRIPTION OF THE DRAWINGS
In the following description of the preferred embodiments, reference is made to the accompanying drawings that form a part hereof, and in which are shown by way of illustration specific embodiments in which the invention may be practiced. It is understood that other embodiments may be utilized and structural changes may be made without departing from the scope of the present invention. The present invention may be practiced according to the claims without some or all of these specific details. For the purpose of clarity, technical material that is known in the technical fields related to the invention has not been described in detail so that the present invention is not unnecessarily obscured. J In the accompanying d rawing: FIG. 1 presents the drug delivery device of the present invention; ] FIG, 2 presents a schematic view of the peritoneum surrounding the body organ; 1 FIGs. 3A-C present a perspective view of the drug delivery device of the present invention; ] FIGs. 4A-B present a cross section view of the drug delivery device of the present invention; ] FIG. 5 presents a cross section view of the drug delivery device of the present invention; ] FIG. 6 presents a cross section view of the drug delivery device of the present invention;
FIGs. 7-12 present a perspective view of a cluster-bomb-like implementable drug delivery systems of the present invention; ana ] FIGs. 13-19 present graphs of the release profile of the drug delivery device of the present invention;
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention have been defined specifically to provide device, means ana method tor delivering a medication to a predefined location at the abdom inal cavity and more particularly to the peritoneal cavity of a patient for a complementary' medical analysis and treatment.
The present invention provides a device, kit and method for localized long-term administration of a therapeutic agent or a medication to a particu lar tissue or organ. T he device can be implanted or inserted once and release variety of drug dosages over an extended period. The present invention provides an implanted device administrating a medication via the intraperitoneal (IP) route in patient's body cavity. Using the IP rout for delivering a medication allows higher medication doses and more frequent administration of drugs, and it is more effective in killing ceils such as cancer cells in the peritoneal cavity, where ovarian cancer is li kely to spread or recur first.
Without wishing to be bound by theory, the peritoneum is known as a membrane that surrounds the visceral organs forming the mesentery which connects the Bowel loops. The physiologic characteristic of the peritoneal cavity provides a useful portal of entry in the body for variety pharmacological agents. Several medications, such as antibiotics can be given via the intraperitoneal (IP) route to treat episodes of peritonitis .The IP route can further be used for delivering a medication in patients with intra-abdominal malignancies, i.e. gynecological and gastrointestinal cancers. The rate and amount of drug transfer in the peritoneum are dependent on several factors. Factors such as peritoneal inflammation, surface area, peritoneal blood flow, time of contact etc., influence the drug transfer. Using the IP route provides high concentration locally which targets the involved peritoneal tissues directly.
Without wishing to be bound by theory, the peritoneum is known to support the abdominal organs and serves as a conduit for their blood and lymph vessels and nerves. The structures in the abdomen are classified as intraperitoneal, retroperitoneal or intraperitoneal depending on whether they are covered with visceral peritoneum and whether they are attached by mesenteries (mensentery, mesocolon). Since the IP fluid surrounds variety of organs such as the stomach, duodenum , jejunum, i leum, cecum, appendix, transverse colon, sigmoid colon, rectum, Liver, spleen, pancreas , uterus, fallopian tubes, ovaries , kidneys, adrenal glands, proximal ureters, renal vessels , urinary bladder and distal ureters, the IP can be an essential route for delivering medication to those organs. The present invention further provides the usefulness of I P drug therapy device and the factors influencing it, as well as strategies to increase the efficacy, and conclude that IP route is an alternate route to the more conventional drug delivery routes, and can be successfu lly used when the target is within the peritoneal cavity or adjacent tissue.
The present invention further provides a device which after introducing into the peritoneal cavity, drug transfer occurs into the surrounding peritoneal tissues, and from there into the body compartments via the circulation.
Peritoneal transport occurs via three simultaneous processes: a) diffusion, b) ultrafiltration, and c) fluid reabsorption. A three pore model has been validated, which suggests that transport across the peritoneal membrane occurs by pores of three different sizes. The large pores and small pores with radius of about <2μ , responsible for the transport of larger and smaller sized solutes. The absorption of intraperitoneailv admin istered macromolecules is linear in time, irrespective of molecular size or concentration.
The transport of the pharmacological agent across the peritoneum is governed by factors such as (a) dosing variables i.e. dose, volume, temperature, duration, composition of carrier solution etc., (b) drug properties i.e. molecular weight, io nic charge, apparent volume of d istribution (Vd), membrane binding, lipid/ water solubility etc, as well as (c) the characteristics of the peritoneum, i.e. surface area, charge, permeability, peritonitis, fibrosis, peritoneal blood flow and lymphatic absorption etc.
The rate and amount of drug transfer are dependent on several factors. The kinetics of the drug, as well as factors such as peritoneal inflammation, surface area, peritoneal blood flow, time of contact, etc. influence the drug transfer. Dedrick et ah, Pharmacokinetic Problems in Peritoneal Drug Administration-, Tissue Penetration and Surface Exposure JNCI ] Nad Cancer Inst (1997) 89(7): 480-487 described in a pharmacokinetic model which showed that with IP infusion the drug concentrations in the peritoneal cavity could theoretica lly exceed those in systemic circulation by three log values or more. Maneuvers such as increasing the dwell duration, temperature or pressure, solutions of different ton icity7, use of vasoactive agents, and surfactants have been tried in an effort to increase the drug efficacy.
The surface area of the peritoneum which actually comes in contact with the therapeutic solution is a small fraction of the total peritoneal surface area. Chagnac et a!., Effect of Increased Dialysate Volume on Peritoneal Surface Area among Peritoneal Dialysis Patients JAS 2002 13(10) 2554-2559 demonstrated that during dialysis, at any given time approximately 30% of the total peritoneal area is covered. For example, newer icodextrin containing solutions can maintain the osmotic gradient for up to 24 hrs or longer, and have been used as a carrier for drugs such as 5FU for up to 96 hours. Other factors such as the solution volume, patient size, and patient position, all affect the contact surface area. An ambulator/ patient will have most of the fluid in the bottom of the cavity which decreases the contact area .There exist anionic charges on the peritoneal basement membrane and capillaries adjacent to it. Changes in these anionic charges can influence transperitoneal absorption of drugs such as aminoglycosides.
The device of the present invention has potential benefits for the use of IP route instead of using other routes such as intravenous (IV) route. The IP route is with the ability to treat the patient on an outpatient basis, presence of an existing access for ad m inistration, avoidance of cost of IV lines, ana avoidance of IV route related toxicities such as phlebitis, and loss of veins which may need to be preserved. For example, in patients with intra-abdominal malignancies, IP route can be successfully used with less systemic toxicity. Furthermore I P route is an alternate manner to deliver a medication, especially where the target is contained within the peritoneal cavity. Gene therapy delivery may further use the IP route. In another embodiment of the present invention, the device is configured to be insertable and/or implanted via natural body orifice or any opening to a cavity or passage of the body. After implantation of the device within the peritoneal cavity, when in IP fluid contact, the drug is released into the surround ing of the peritoneal tissues and from there into the body compartments via the circulation. T he med ication rate and amount released are dependent on several factors such as the kinetics of the drug, peritoneal inflammation, surface area, peritoneal blood flow and time of contact, the medication adm inistrated vi the IP route provides high concentration locally of the medication which further targets the involved peritoneal tissues directly. The major advantage of IP therapy is regional dose intensity. After intracavitary drug administration, the peritoneal cavity is exposed to higher concentrations compared to other parts of the body. The concentration differential occurs because drug movement from peritoneal cavity7 to plasma (peritoneal clearance) is generally slow relative to drug clearance from the body.
In another embodiment of the present invention, the peritoneal cavity is the site of disease in several cancers, including ovarian, gastrointestinal and peritoneal mesothelioma. In another embodiment of the present invention, Endometriosis, a gynecological medical condition in which cells from the lining of the uterus (endometrium) appear and flourish outside the uterine cavity, most commonly on the membrane which lines the abdominal cavity. The uterine cavity is lined with endometrial cells, which are under the influence of female hormones. Endometrial-! ike cells in areas outside the uterus (endometriosis) are influenced by hormonal changes and respond in a way that is similar to the cells found inside the uterus. Symptoms often worsen with the menstrua l cycle.
Without wishing to be bound by theory, Endometriosis is typically seen during the reproductive years. Symptoms may depend on the site of active endometriosis. Its main but not universal symptom is pelvic pain in various manifestations. Endometriosis is a common finding in women with infertility. Endometriosis can be treated in a variety of ways, includ ing pain medication, hormonal treatments, and surgery. In Endometriosis disease endometrial glands and stroma implant and grow in areas outside the uterus. The location a nd inflammatory response to these lesions are believed to play key role in symptoms ana signs associated with endometriosis. The present invention provides an device which enables a medication delivery to a target region, organ or area related and affected of Endometriosis disease within the body cavity of a patient.
The device of the present invention may be further provided for the treatment of Endometriosis and for variety types of Cancer such as Ovarian, Peritoneal, Pseudomyxoma peritonei, Mucinous, adenocarcinoma of the appendix, Mesothelioma and Colorectal carcinoma. The device may further be provided for treating variety of Infection such as: Peritonitis, P! L) - acute, PID - chronic and Tuberculosis.
The device may further be provided for treating Familial Mediterranean fever (FMF) ana for prevention of adhesions: Post-operative and Post-infectious. The device may further be provided for the treatment of extraperitoneal pathology: such as Inflammatory Bowel Disease- IB D (Crohn's disease and Ulcerative colitis) and reservoir for chronic system ic treatment.
The term "controlled release system" refers hereinafter to but is not limited to , drug delivery- systems (DDS), controlled release systems, delayed release systems, modified release systems, slow release systems, pH dependent systems, pH independent coatings systems, and any combinations thereof. It is further in the scope of the invention, wherein the term "controlled release " refers hereinafter to but is not limited to sustained release, sustained action, controlled release, extended action, timed release dosage forms and other parallel terms used to identify drug delivery systems that are designed to achieve a prolonged therapeutic effect by continuously releasing medication over an extended period of time after the administration of single dose.
In reference to "Concept and system design of rate-controlled drug delivery system" a presentation held by Basavaraj K. Nanjwade (05/10/2010), currently available at http://api.ning.com/files/CEij lTrVJQblwjA8jyH mSCx4r lR52s091 EoSCX4iPCCc8mGmeK UZQTKByLUBS6hAVA3HPAN8hN ILNIRLI UuJFVTWgVaTEQru/ConceptandSystemDesig nforRateControlledDrugDelivery.ppt, there are various types of controlled release systems, such as physically enabled systems, chemically enabled systems, biochemically enabled systems etc., all of wh ich are incorporateable within the present invention and technology thereof.
Physically enabled systems are selected, in a non-limiting manner, from osmotic pressure- activated DDS; hydrodynamic pressure-activated DDS; vapor pressure-activated DDS; Mechanically activated DDS; magnetica lly activated DDS; mono-phoresis activated DDS; iontophoresis activated DDS; hydration-activated DDS etc. Chemically enabled systems are selected, in a non-limiting manner, from pH- activated DDS; ion- activated DDS; hydrolysis- activated DDS etc. Biochemically enabled systems are selected, in a non-limiting manner from enzyme- activated DDS and other biochemical- activated DDS.
In one embodiment of the invention, an implemeritable drug delivery pump (DDP) is provided useful for facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body. The DDP comprises a. at least one envelope with at least first layering surface for containing a medication; the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the IP by the fluid; and at least one first volume confined by the envelope, comprising effective measure of the med ication; and a medication delivery pump; wherein the medication delivery pump facilitates the delivery of the medication from the at least one envelope.
In one embodiment of the invention, a method of facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body is provided useful. The method comprises a step of providing an implementable drug delivery pump (DDP) by containing a medication within at least one envelope with at least first layering surface; providing the envelope with at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the I P by the fluid; a step of confining by the envelope at least one first volume, comprising effective measure of the medication; and a step of facilitating the delivery of the medication from the at least one envelope by means of a medication delivery pump.
It is thus well within the scope of the invention wherein the DDS are in connection with or comprises drug's or fluid's pump such that a DDP is provided. Hence for example, an Alzet® - like osmotic pump is utilized. Com mercially available ALZET osmotic pumps and their like are miniature, infusion pumps for the continuous dosing. Alternatively or additionally another DDP is provided, namely a hydrodynamic pressure-activated drug delivery system (e.g., push- pull osmotic pump) is utilized. This system is fabricated by enclosing a collapsible, impermeable container, which contains liquid drug formulation to form a drug reservoir compartment inside rigid shape-retaining housing. A composite laminate of an adsorbent layer and a swellable, hyd roph ilic polymer layer is sandwiched. Alternatively or additionally another DDP is provided, namely a vapor pressure-activated drug delivery system is utilized. In this system, the drug reservoir in a solution formulation is contained inside an infusate chamber. It is physically separated from the vapor pressure chamber by a freely movable bellows. The vapor chamber contains a vaporizabie fluid, which vaporizes at body temperature and creates a vapor pressure. Under the vapor pressure created, the bellows moves upward & forces the drug solution in the infusate chamber to release, through a series of flow regulators and delivery cannula into the blood circulation at a constant flow rate. Alternatively or additionally another DDP is provided, namely a mechanically activated drug delivery system, is utilized. In this type, drug reservoir is in solution form retained in a container equipped with mechanically activated pumping system. Alternatively or additionally another DDP is provided, namely magnetically activated drug delivery system is utilized. In this type, drug reservoir is a dispersion of peptide or protein powders in polymer matrix from, which macromolecular drug can be delivered only at a relatively slow rate. ' Ibis low rate of delivery can be improved by incorporating electromagneticaliy triggered vibration mechanism into polymeric device combined with a hemispherical design. Device is fabricated by positioning a tiny magnet ring in core of hemispherical drug dispersing polymer matrix. Device is fabricated by positioning a tiny magnet ring in core of hemispherical drug dispersing polymer matrix. T he external surface is coated with drug impermeable polymer (ethylene vinyl acetate or silicon elastomer) except one cavity at the center of the flat surface. This delivery device used to deliver protein drugs such as bovine serum albumin, at a low basal rate, by a simple diffusion process under non triggering condition, As the magnet is activated to vibrate by external electromagnetic field, drug molecules are delivered at much higher rate. Alternatively or additionally another DDP is provided, namely sonophoresis - activated drug delivery system (phonophoresis) is utilized. This type of system utilizes ultrasonic energy to activate or trigger the delivery of drug from polymeric drug delivery device. System can be fabricated from non-degradable polymer (ethylene vinyl acetate) or bioerodiable polymer (poly[bis(p-carboxyphenoxy) alkane an hydride. Alternatively or additionally, iontophoresis activated drug delivery system is utilized. This type of system uses electrical current to activate and to modu late the diffusion of charged drug across biological membrane. Alternatively or additionally another DDP is provided, namely hydration activated drug delivery system is utilized, in this system, the drug reservoir is homogeneously dispersed in a swellable polymer matrix fabricated from a hydrophilic polymer (ethylene glyxomethacrylate). The release of drug is controlled by the rate of swelling of polymer matrix. Alternatively or additionally, a pH- activated drug delivery system is utilized. This type of chemically activated system permits targeting the delivery of drug only in the region with selected pH range. It fabricated by coating the drug-containing core with a pH - sensitive polymer combination. For instances, a gastric fluid labile drug is protected by encapsulating it inside a polymer membrane that resist the degradative action of gastric pH. Alternatively or additionally another DDP is provided, namely ion- activated drug delivery system is utilized. An ionic or a charged drug can be delivered by this method and this system are prepared by first compiexing an ionic drug with an ion-exchange resin containing a suitable counter ion. Hence for example, by forming a complex between a cationic drug with a resin having a SO 3 group or between an anionic drug with a resin having a N(CHi)3 group. The granules of drug-resin complex are first treated with an impregnating agent & then coated with a water-insoluble but water-permeable polymeric membrane. This membrane serves as a rate- controlling barrier to modulate the influx of ions as well as the release of drug from the system In an electrolyte medium, such as gastric fluid ions diffuse into the system react with drug resin complex & trigger the release of ionic drug. Since the GI fluid regularly maintains a relatively constant level of ions, theoretically the deliver/ of drug from this ion activated oral drug delivery system can be maintained at a relatively constant rate. Alternatively or additionally another DDP is provided, namely hydrolysis- activated drug delivery system is utilized. This type of system depends on the hydrolysis process to activate the release of drug. Drug reservoir is either encapsulated in microcapsules or homogeneously dispersed in microspheres or nano particles tor injection. It can also be fabricated as an implantable device. All these systems can be prepared from bioerodib!e or biodegradable polymers (polyanhydride, polyorthoesters). It is activated by hydrolysis-induced degradation of polymer chain & is controlled by rate of polymer degradation. Alternatively or additionally another DDP is provided, namely an enzyme - activated drug delivery system is utilized. This type of biochemical system depends on the enzymatic process to activate the release of drug. Drug reservoir is either physically entrapped in microspheres or chemically bound to polymer chains from biopolymers (albumins or polypeptides). The release of drug is activated by enzymatic hydrolysis of biopolymers (albumins or polypeptides) by specific enzyme in target tissue. Alternatively or additionally another DDP is provided, namely a feedback regulated drug delivery system is utilized. In this group the release of drug molecules from the delivery system is activated by a triggering agent. Rate of drug release is control led by concentration of triggering agent. Alternatively or additiona lly another DDP is provided, namely bioerosion-regulated drug delivery system is utilized. The system consisted of drug-dispersed bioerodible matrix fabricated from poly (vinyl methyl ether) ester which is coated with layer of immobilized urease. In a solution with near neutral pH, the polymer only erodes very slowly. In presence of urea, urease metabolizes urea to form ammonia. This causes increase in pH & rapid degradation of polymer with release of drug molecule. Alternatively or additionally, bioresponsive drug deliver/ system. Drug reservoir is contained in device enclosed by bioresponsive polymeric membrane whose drug permeability is controlled by concentration of biochemica l agent. A lternatively or additional ly another DDP is provided, namely a self-regulating drug delivery system is utilized. This type of system depends on a reversible & competitive binding mechanism to activate and to regulate the release of drug. Drug reservoir is drug complex encapsulated within a semi permeable polymeric membrane. The release of drug from the delivery system is activated by the membrane permeation of biochemical agent from the tissue in which the system is located.
The term "unit dosage form" or "dose form" or "drug" or "medication" or "therapeutic agent" interchangeably refers hereinafter to a pharmaceutically acceptable forms of a liquid, gel, solid, gas or semisolid dosage forms drug selected from the group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates, Emulsion, Hydrogel, Molecular encapsulation, Soitgel, Solution, Suspension, Syrup, Tincture, Tisane or any combination thereof, in a sustained-release formulations.
The core of the unit dosage form, may be coated with at least one functional coating or film which may modify the release properties of the formulation. The unit dosage form of the present invention may further comprise d ifferent drug release profiles from d ifferent layers. I'he medications can be any compound that is biologically active and requires long term ad m inistration to a tissue or organ for maximum efficacy. Therapeutic agents that can be used in accordance with the present invention include, but are not limited to, antimuscarinic agents, anticholinergic agents, antispasniodic agents, calcium, antagonist agents, potassium, channel openers, musculotropic relaxants, antineoplastic agents, polysynaptic inhibitors, and beta- ad renergic stimulators. Examples of anticholinergic agents are propantheline bromide, imipramine, mepenzolate bromide, isopropamide iodide, clidinium bromide, anisotropine methyl bromide, scopolamine hyd rochloride, and their derivatives. Examples of antimuscarinic agents include, but are not limited to, hyoscyamine sulfate, atropine, methantheline bromide, emepronium. bromide, anisotropine methyl bromide, and their derivatives. Examples of polysynaptic inhibitors are baclofen and its derivatives. Examples of 8-adrenergic stimulators are ter-butaline and its derivatives. Examples of calcium antagonists are terodiline and its derivatives. Examples of musculotropic relaxants include, but are not limited to, dicyclomine hydrochloride, flavoxate hydrochloride, papaverine hydrochloride, oxybutynin chloride, and their derivatives. Examples of an antineoplastic agents include, but are not limited to, carmusti ne levamisole hyd rochloride, flutamide, (w-methy!-N-[4-nitro-3-(tri.fluorom.ethy!) phenyl]), adriarnycin, doxorubicin hydrochloride, idamycin, fluorouracil, Cytoxan, mutamycin, mustargen and leucovorin calcium..
In another embodiments of the present invention, the device may contain medication in a gas form (e.g Nitric Oxide,NO) such that when the inner structure of the device is in contact with the I P fluid a gas material is released. The gas material may further provide antimicrobial, anti- infection, and/or therapeutic activity and characteristics.
In. another embodiment of the present invention, one of the uses for the control release delivery' of drugs to a specific tissue or organ is for internal organs that can be classified as mechanically dynamic organs. Examples of mechanically dynamic organs include, but are not limited to, the stomach, intestines, heart, bladder, ureter, urethra, various sphincter muscles, and the esophagus. The functions of these organs are maintained by their normal and timely mechanical contractions. Should the mechanical properties be lost, the intended functions of these organs are partially or completely lost- Reference is now made to Fig. 1 , which illustrates a biocompatible drug delivery device 1 for the delivery of at least a first med ication and at least second med ication to a patient body cavity, comprising: at least one first chamber 3 and at least one second chamber 4 for loading at least one first med ication and at least one second medication respectively, (b)at least one drug- releasing component 2 adapted for medication release; and optionally, (c)an anchoring means 6 adapted for securing the device to a body tissue. The drug-releasing component is adapted for controlling the rate of release of the medication from the device internal surface or external surface. The medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body.
The present invention further provides an implementable drug delivery device (DDL)) for delivering a medication via the intraperitoneal (I P) to a target region in a patient body, the DDD comprising:(a)at least one envelope with at least first layering surface for containing a medicamen; and, (b)at least one first volume confined by the envelope, comprising effective measure of the medication; wherein the envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the medication towards the I P by the fluid.
The present invention further provides a drug delivery device (DDD) for activating and delivering at least one activated medication via the intraperitoneal (IP) route to a target region in a patient body, the DDD comprising: (a) a hollow container having an external surface and internal su rface for housing an inactive medication, the inactive med ication renderable active by contact with the IP fluid; (b) at least one first chamber for loading the medication into the hollow container; and, (c) at least one second chamber for releasing the at least one activated medication into the interior body cavity; wherein the external surface is provided with a functional separation element (FSE) having least one protuberance or/ and indentation for distancing the hollow container from interior walls of the body cavity; further wherein the first and second chambers provide an ingress path for IP fluid to enter the hollow conta iner and an exit path for expulsion of activated medication. The present invention further provides a biocompatible drug delivery device for the deliver/ of at least one medication to a target region within patient body cavity, comprising: (a) at least one first chamber and at least one second chamber for containing at least one first medication and at least one second medication respectively? (b) at least one drug-releasing component adapted for medication release, located between the chambers, and optiona lly, (c)an anchoring means adapted for securing the device to a body tissue. The each of the chambers are easily connected or disconnected from the device when loading a medication especial ly in surgical or emergency procedure.
The med ication is delivered first to the intraperitoneal (IP) route second to a first target region and third to a second target region. The drug-releasing component is adapted for controlling the rate of release of the medication when in IP fluid connection.
Furthermore the drug transfer via the I P route achieves therapeutic efficacy in the region of interest while minimizing the systemic toxicities.
The present invention further provides an intraperitoneal (I P) administrable device useful for treating endometriosis, the device comprising: at least a first chamber and a second chamber for containing the at least a first medication and at least a second medication respectively; and , an anchoring means adapted for securing the device to a body tissue; wherein the device is configured for controlling the rate of release of the medication when the medication is delivered to a target region via the intraperitoneal (IP) route .
The drug-releasing component may be a selective barrier element (not shown) attached to at least first chamber containing at least one medication. The separating element may be a membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through, but not other. T he selective barrier shape and structure may further control the time of release of each medication within each chamber.
The medication may further be based upon a controlled release delivery system of the medication. The controlled release delivery system may be a slow release, immediate release, sustained release and combination thereof.
The drug-releasing component may further be a combination of a separating element and a controlled release delivery system of at least first medication. The drug delivery device may be composed of a biodegradable material selected from the group consisting ot: liposomes, polylactides (polylactic acid), polyglycolide (polymer ot glycolic acid), poly (lactide co-glycolide (co-polymers ot lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chonclroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone container.
In another embodiment of the present invention, the drug delivery device contains at least first medication which is physically trapped, or covalently or ionically immobilized in a biodegrad able matrix.
In another embodiment of the present invention, the device is adapted for loading a medication having a dosage form selected from a group consisting ot aliquot, tablet, caplet, capsule, pi ll, bolus, particle, micronized particle, particulate, pellet, core, powder, granu le, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof. The dosage form is further based upon controlled release delivery system. The controlled release delivery; system may be a slow release, immediate release, sustained release and combination thereof. T herefore at least first medication is based upon a slow release manner whilst at least second medication is based upon immediate release manner. Furthermore, at least first medication is based upon a slow release manner whilst at least second medication is based upon sustained release manner
In another em bodiment of the present invention, the device is a 3D container having a cylinder shape, a tube like, a star like, a ball like, a roll like. The device is composed of a semi permeable wall having at least one opening channel for IP fluid entrance and medication release. Furthermore the device is a non-absorbable, flexible container having an elongated shape.
In another embodiment of the present invention , the device is adapted to release at least first medication at a rate of at least approximately 0. 1 meg day whilst at least second medication is released at a rate of at least approximately 0.5 meg/day over a period of at least approximately one week. The medications may be transported via a) diffusion, b) ultrafiltration, c) fluid re- absorption and combination thereof. The device may contain medications for treating at least one diseases, disorder or condition having different dosages for long treatment. In another embodiment of the present invention, the device is used for treating Endometriotic disease.
The device of the present invention is a volume-adjustable device. Thereby it may be adjusted to the volume or number or medication elements contained within the medication delivery device. The medications for IP delivery for treating of endometriosis are selected in a non- limiting manner from the group consisting of Progestatives - e.g. dienogest, Progesterone antagon ists / modulators such as mifepristone, GN RH agon ists - e.g. leuprolide acetate, Aromatase inhibitors - e.g. letrezo!e, NTHEs - e.g. parecoxib, Anti-inflammatory', Anti- angiogenic DA - e.g. cabergoline, N F Kappa-b medications, Vitamin D, Estrogen and a combination or mixtures thereof.
In another embodiment of the present invention, the device may Last and provide treatment within patient's body cavity tor at least a week.
In another embodiment of the present invention, the device may comprise an anchoring means for maintaining the device with in the body cavity. The device is flexible and shaped to be insertable and/or implanted into the body IP cavity. The device may be preferably liquid impermeable or at least liquid sem i-permeable. The device further comprises at least one opening through which the medication can be employed and/or release. Preferably, the medication is situated and released from at any one of the following locations: a. the external surface of the device, b. the internal surface of device within at least one of the chambers and, c. within a network of pores of wh ich the device may comprise.
In another embodiment of the present invention, the regions within the body cavity may be chosen from the group consisting of. rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra (including prostate), bladder and, intra-articuiar cavity. The device is capable of deliver/ for local treatment and/or systemic treatment.
The device of the present invention may combine several medications in order to increase the therapy effect in several body regions or organs and further reduce side effects.
In another embodiment of the present invention, the device may contain variety of substance combinations, shapes or presentations (capsules, tablets, soft gels) without chemical interaction. The device of the present invention provides controlled release of drug in vivo by diffusion of the drug out of a polymer and/or by degradation of the polymer over a predetermined period following administration to the patient.
In another embodiment of the present invention, the medication may be employed into the implanted device using a syringe or catheter.
In another embodiment of the present invention, the device may additionally comprise an applicator for affixing the device to the body tissue via anchoring means. The applicator may further be used for reloading the device chambers with additional medications. The applicator may further be used tor better implantation ot the device within the body cavity of the patient. The applicator is configured to enable the device to collapse easily into the body cavity.
Reference is now made to Fig 2 which is a prior art, presenting the peritoneal cavity. The peritoneum is a membrane that forms the lining of the abdom inal cavity or the coelom it covers most ot the intra-abdominal (or coelomic) organs in amniotes ana some invertebrates (annelids, for instance). It is composed of a layer of mesothelium supported by a thin layer of connective tissue. The peritoneum both supports the abdominal organs ana serves as a conduit for their blood and lymph vessels and nerves.
The structures in the abdomen are classified as intraperitoneal, retroperitoneal or intraperitoneal depending on whether they are covered with visceral peritoneum and whether they are attached by mesenteries (mensentery, mesocolon).
Reference is now made to Figs. 3a-c which illustrate the configuration of a biocompatible drug delivery device for activating and delivering at least one activated medication via the intraperitoneal (IP) route to at least one target region in a patient body, comprising: (a)a hollow container having an external surface and internal surface for housing an inactive medication, the inactive med ication renderable active by contact with the IP fluid, (b) at least one first chamber tor loading the at least one inactive medication into the hollow container, and (c)at least one second chamber for releasing the at least one activated medication into the interior body cavity; wherein the external surface is provided with a functional separation element (FSE) having least one protruberance or/ and indentation for distancing the hollow container from interior walls ot the body cavity the FSE; further wherein the chambers provide an ingress path for IP fluid to enter the hollow container and an exit path for expulsion of activated medication. In another embodiment of the the present invention, a biocompatible drug delivery device for the delivery of at least one first medication to patient's body cavity via the intraperitoneal (IP) route to at least one target region in a patient body, comprising: an implant for housing and releasing the medication, wherein the implant further comprises at least one chamber for containing the drug; the implant is adapted for introduction and placement into the Peritoneal cavity via the Douglas pouch, the belly-button or any other natural body enter. .
As used herein, the interior walls referees to the device walls, the interior surface of the device or/ana the central surface of the device.
The release of the medication from the implant may be by an actuation element selected from a group consisting of: IP fluid water, IP fluid electrolytes, I P fluid antibodies, I P fluid white blood cells, IP fluid bio-chemicals or any of the martials which the IP fluid consist.
The device may7 further comprise a shielding material (not shown) as an external coating surrounding the device. The shielding is adapted as a barrier or a septum to prevent contact of the device outer surface with the body tissue and furtherniore preventing increment of the concentration release of the medication. Ί he shielding material may comprise plurality of holes.
Ί he device of the present invention may be inserted via the Douglas pouch, rectum, vagina or any other natural body enter.
The device of the present invention may7 further be inserted placed and transported via other fluidly routes or humid regions such as the eye area, the digestive system, the bladder, the vagina or the like. In a nother embodiment of the present invention, Fig 3c illustrates a cylindrical body configuration of the device of the present invention having an external surface comprising a plurality7 of protrusion such as actuation flaps or pins allowing the movement of the device within the body cavity and further allowing the device transported with the IP fluid flow7 to wonder around the organs. The pins allow7 movement of the device in the IP fluid and further to deliver at least one medication to a targeted region.
In another embodiment of the present invention, the device may comprise a monitor unit or a detecting unit for measuring physical or/and chemical characteristics or parameter with the device and fluid conditions such as temperature, humidity, pressure, pH, ionic change, osmolarity, concentration of substances such as acid-lactic, glucose, lipids, hormones, gases, enzymes, inflammatory mediators, plasmin, albumin, lactoferrin, creat.in.in, proteins and the like. The monitor may be programed to begin or stop the release of the medication according to a pre-programed range of at least one of the mentioned parameters. The monitor may further be programed to instruct and control the medication dosage release from the device.
In another embodiment of the present invention, the device may further comprise anchoring means for securing the device implanted to a wall of an organ within a patient body cavity. The anchoring means may be located in the distal portion or the proximal portion of the device.
Additionally, the illustrated configuration advantageously increases flexibility of the device and allow the device to be more effectively navigated through the IP fluid without damaging the device or injuring the patient tissues or organs.
Reference is now made to Fig. 4a-h which illustrates the inner surface of the biocompatible drug delivery device for the delivery of at least one first medication to patient's body cavity. The device has an inner configuration comprising plurality of pores 42 providing a control release mechanism for the medication within the device. The inner surface has a resilient structure perforated by pores through which the drug may be delivered from the chambers of the device.
The medicament may further be placed in any location or side within the device such that when the device is in fluid contact with the I P fluid the medication is released.
In another embodiment of the present invention, the device when placed in contact, with the patient's organ or I P fluid, provides active delivery of at least one medication into the IP fluid. Furthermore the pores may be expandable for enhancing the diffusion and the medication release from, the device.
The outer surface of the devise may comprise severa l layers having pores in different diameters in order to control the rate of the medication release from the device. The device may comprise a first outer layer having pores with smal ler pore diameter and a second layer below the first layer having larger pore diameter for controlling the drug release from the device.
The drug may further be d ispersed in the polymer and re leasable when, the intra surface of the device is in contact with a liquid. The rate of drug release may be dependent on the particular porous structure upon at least one outer layer of the device, which can be specifically formed to achieve a desired rate of release. The pores may have round or other cross-sectional shapes and may have different, sizes. As used herein, a pore diameter refers to die average or ettective diameter of die cross- sections ot the pores. Ί he effective diameter of a cross-section that is not circular equals the diameter of a circular cross-section that has die same cross-sectional area as that of t e non-circular cross- section.
In other embodiments of the present invention, the pores may be filled with peritoneal fluid; the sizes of the pores may change dependi ng on the peritoneal fluid content in the device. The. outer layer may thus behave like a sponge. The pore diameter may be in the range from about 10 to 1 00 nm such that when the outer layer is in a dry condition wherein the peritoneal fl id content of the outer layer is at or near minimum .Furthermore, while a larger pore diameter may provide taster release initially, a smaller pore d iameter with increased lengt of interconnected pores can provide a more steady release at a similar rate. The release rate may also be affected by the manner in which, the drug is incorporated in the device surface. When the drug is initially located in the device matrix, the rate may be slower. When the drug is initially located i the fluid, the rate may be higher. Thus, the drug delivery rate may be controlled by controlling the pore structure and the manner in which the drug is incorporated in the device.
The diameter of the pores 41 may be between a 1 micrometer to about 1000 micrometers (1 mm), allowing exit of the medication in a dosage adjacent to the desired treatment.
Reference is now7 made to Fig. 5 which illustrates the cross section, of the external surface and the internal surface of the biocompatible drug delivery device for the delivery of at least one first medication to patient's body cavity. The device may be composed of at least one medication or having different layer of several medications or different dosages of the same medication.
The internal surface of the device may be made of at least one medication 52 as an inner layer. The device may further contain plurality of pores 52 allowing the entrance of the IP fluid and exit of the medication dissolved in the IP fluid. Fig 5 further illustrates the outer layer of the device comprising plurality of pins adapted for preventing contact with the body tissue.
Fig 5 further illustrates the outer surface comprising plurality of protrusions having a pin like shape adapted to prevent any contact and further to distance the device from the patient's body tissue.
4.1 Reference is now made to Fig, 6 which presents a cross section view oi the biocompatible drug delivery device for the delivery of at least one medication to patient's body cavity. The device comprises an outer shielding comprising plurality of pores 60 adapted to control the medication release from the device and in parallel preventing contact with the body tissue. The device of Fig. 6 has a tubular configuration comprising an inner hollow tubu lar 61 structure allowing entrance of the IP fluid and an inner channel for loading at least one medication 62. T he device configuration allows the entrance of the I P fluid and exit of the medication dissolved in the IP fluid. The medication is released when in fluid contact with the IP fluid.
In another embodiment of the present invention, the device may be permeable or semipermeable to the IP fluid such that the IP fluid transports the med ication to a target region. The device outer shielding prevents physical contact with the body tissue attached or anchored to the device and therefore preventing medication release with in the tissue. The I P medication transport may occur via. pores having d ifferent sizes located in the outer shield ing of the device. The absorption of intraperitoneally administered medication is linear in time, irrespective of molecular size or concentration.
In another embodiment of the present invention, the device of the present invention provides minimal fluctuations caused by temperature, humidity, pressure, pH, ionic change, osmolarity, concentration of su bstances such as acid-lactic, glucose, lipids, hormones, gases, en zymes, inflammatory mediators, plasmin, albumin, lactorerrin, creatinin, proteins and the like. The device is further Light weight and configured to prevent any friction with a surrounding tissue more particularly when the device is placed between organs.
In another embod iment of the present invention the device is easily inserted and i n parallel easily retrieved. The device is biocompatible and hypoallergenic therefore does not cause any side-effects.
In another embodiment of the present invention the device is radiopaque therefore is opaque to one or another form of radiation, such as X-rays. The device may further block radiation rather than allow it to pass through. Table 1 below presents a variety of regions in the peritonea ! cavity which the device can be located in and further release a medication.
Intraperitoneal Retroperitoneal Infraperltoneal /
Subperitoneal
Stomach, hirst part of the duodenum, The rest of the duodenum, Rectum
jejunum, ileum, cecum, appendix, ascending colon, descending
transverse colon, sigmoid colon, colon, rectum
rectum
Liver, spleen, pancreas Pancreas
Kidneys, adrenal glands, Urinary bladder, distal proximal ureters, renal ureters
vessels
uterus, fallopian tu bes, ovaries Gonadal blood vessels
Inferior vena cava, aorta
The present invention further provides a method for admi nistering a dosage form to a patient, comprising steps of (a) providing a biocompatible drug delivery device for the delivery ot at least a first medication and at least second medication to a patient body cavity, comprising: (i)at least a first chamber and a second chamber for containing the at least one first medication and at least one second medication respectively, (iii) at least one drug-releasing component adapted for medication loading and release; optionally, (iv)an anchoring means adapted for securing the device to a body tissue; (b) inserting the drug delivery device to a body cavity of the patient, (c) optimally anchoring the drug delivery device to the body tissue of the patient; (d)releasing of the dosage form via drug-releasing component of the device into a the body cavity of the patient body.The releasing of a dosage form via drug-releasing component is associated with the device internal surface or/and external surface for controlling the rate of release of the medication.
The method of the present invention further comprising the step of delivering the medication via the intraperitoneal (I P) route to at least two target regions in a patient body.
The present invention further provides a method of manufacturing a biocompatible drug delivery' device for the delivery of at least a first medication and a second medication to a patient body cavity, comprising steps or: (a) providing a container containing at least a first chamber and a second chamber for containing the at least a first medication and the second medication respectively; (b) attaching anchoring means to at least one edge of the container; (c) connecting at least one drug-releasing component to at least one first chamber, the drug- releasing component is adapted for medication loading and release, and (d) providing a drug- releasing component for medication release. The drug-releasing component is associated with the device internal surface or/ and externa! surface for controlling the rate of release of the medication via the intraperitoneal (IP) route to at least two target regions in a patient abdominal cavity.
It is one object of the present invention to provide a method for administering a drug delivery device to a target region within a patient body cavity, comprising steps of: (a) providing a biocompatible drug delivery device for the delivery of at least a first medication and a second medication to the intraperitoneal (IP) route, comprising: (i)at least one opening for loading and releasing of at least a first med ication and at least one second medication; (v)a drug- releasing component for medication release, and (vi) an anchoring means adapted for securing the device to patient body tissue; (b) provid ing an applicator; (c) placing the drug delivery device within the end of the applicator; (d) inserting the end of the applicator to target region within patient body cavity; (e) releasing the device from the end of the applicator whereby the device automatically seats itself within the patient body cavity, ana (0 withdrawing the applicator from the patient body cavity; wherein the method comprising the step of delivering the first medication and second medication via the intraperitoneal (IP) route to at least two target regions in a patient body.
It is another object of the present invention to provide method for administering a medication to a patient, comprising steps of: (a)providing biocompatib le drug delivery device for activating and delivering at least one activated medication via the intraperitoneal (IP) route to at least one target region in a patient body comprising: (i)a hollow container having an external surface and internal surface for housing an inactive medication, the inactive medication renderable active by contact with the IP fluid, (ii)at least one first chamber for loading the at least one inactive medication into the hol low container; and (iii)at least one second chamber for releasing the at least one activated medication into the interior body cavity, (b) introducing the drug delivery device to the patient's body cavity. The device may be introduced via natura l body entrance, by injection procedure or by a surgical procedure. In another embodiment of the invention the method as described above, wherein the releasing the medication is from a first configuration suited for passage of the drug delivery device through the drug-releasing component to a second configuration suited for retaining the drug delivery device with the intraperitoneal (IP) direction route.
In another embodiment of the invention the method as described above, wherein the medication dosage form is selected from a group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
In another embodiment of the invention the method as described above, wherein the method comprising the step of providing a medication in a liquid phase, gas phase or solid phase.
In another embodiment of the invention the method as described above, wherein the method comprising the step of providing a medication based upon a controlled release delivery system.
In another embodiment of the invention the method as described above, wherein the controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
In another embodiment of the invention the method as described above, wherein the device is a biodegradable container containing the medication that has been physically trapped, or covalently or ionically immobilized in the biodegradable matrix.
In another embodiment of the invention the method as described above, wherein the biodegradable is selected from the group consisting of: liposomes, po!y!actides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycoltde (co-polymers of lactic acid and glycolic acid) po!yanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
In another em bodiment of the invention the method as described above, wherein the container is a cylinder or tube like implanted device.
In another em bodiment of the invention the method as described above, wherein the device is a non-absorbable, flexible container having an elongated shape.
In another embodiment of the invention the method as described above, wherein the method comprising the step of releasing the at least first medication at a rate of at least approximately 0.1 meg/day and at least second medication at a rate of at least 0,5 meg/day over a period of at least approximately one week.
In another embodiment of the invention the method as described above, wherein the device is used for treating Endometriotic d isease.
In another em bodiment of the invention the method as described above, wherein the device is semi permeable.
In another embodiment of the invention the method as described above, wherein the regions are selected from the group consisting of: rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity, eye and combination thereof.
In another embodiment of the invention the method as described above, wherein the method comprising the step of transporting the medication to the Peritoneal fluid via a) diffusion, b) ultrafiltration, c) fluid re-absorption and combination thereof.
In another em bodiment of the invention the method as described above, wherein additionally comprising step of affixing and securing the device to the body tissue using an applicator via anchoring means.
The present invention further provides a kit for the delivery of at least a first medication and a second medication to a patient body cavity, comprising: (a)a biocompatible drug delivery device comprising: (i) at least a first chamber and at least second chamber for loading and releasing the at least a first medication and the second medication respectively, (ii) optionally an anchoring means adapted for securing the device to a body tissue; and (iii) a drug-releasing component for controlling the rate of release of the medication, (b) an applicator adapted for affixing the device to the body tissue via anchoring means.
In another embodiment of the invention the device can travel and wander around the peritoneal cavity without any anchoring means.
In another embodiment of the invention, at least first medication is delivered via the intraperitoneal (IP) route to at least two target regions in a patient body.
In another embodiment of the invention the kit as described above, wherein the applicator is adapted for inserting the device into the body or an individual or/and for removing the device from the patient.
In another embod iment of the invention the kit as describee! above, wherein the applicator is further adapted tor reloading the first medication and/or second medication into the device cham bers.
In another embodiment of the invention the kit as described above, wherein the kit is useful for treating endometriosis.
In another em bodiment of the invention, an implementable drug delivery device (DDD) is provided useful for delivering a medication via the intraperitoneal (IP) to a target region in a patient body. The DDD comprises at least one envelope with at least first layering surface confining at least one first volume accommodating at least one first precursor and at least one second precursor. The envelope comprises at least one ingress path for IP's fluid to both inlet and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid. At least one first precursor and at least one second precursor forms an effective medication to target the region in a patient body.
In another embodiment of the invention, a method of targeting a region in a patient body is disclosed. The method comprises steps as follows: providing a drug delivery device (DDD) with at least one envelope, comprising at least first layering surface; confining within the envelope at least one first precursor and at least one second precursor; providing within the envelope at least one ingress path tor I P's fluid to both in let and outlet the layer for expulsion of the first and second precursors towards the IP by the fluid; forming, by means of the at least one first precursor and at least one second precursor, an effective medication; and targeting the newly formed medication the region in a patient body.
It is in the scope of the invention wherein the at least one first and at least one second precursors forms the medication(s) by reaction, polymerization, condensation, oxidation and reduction, complexation, acid-base reactions, precipitation, solid-state reactions, photochemical reactions, substitution, addition and elimination, rearrangement reaction, biochemical reactions etc.
It is in the scope of the invention wherein the at least one first and at least one second precursors forms the medication(s) within the DDD, outside the DDD, adjacent the targeted organ, on the targeted organ etc.
It is in the scope of the invention wherein a feedbacked implementable drug delivery device (FDDD) is provided useful for delivering a time-resolved dose of one or more medications via the intraperitoneal (IP) to a target region in a patient body.
The FDD D comprises e.g., at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; the envelope comprises at least one controllable path for I P's f lu id to both inlet and outlet the layer for controlled expulsion of the medication towards the IP by the fluid; and a fluid's flow control ling means; wherein the controlling means regulates the path thereby controlling the fluid flow.
It is in the scope of the invention wherein in the FDDD defined above, the fluid flow is selected from a group consisting of IP's fluid inflow, the medication's outflow or a combination of the same.
It is in the scope of the invention wherein in the FDDD defined above, the device further comprises a sensing means in communication with the controlling means, for analyzing one or more mem bers of a group consisting of IP's parameters and targeted region of the body, thereby controlling the fluid flow in a feedbacked manner.
It is also in the scope of the invention wherein a method of delivering a time-resolved dose of one or more medications via. the intraperitoneal (IP) to a target region in a patient body is provided usefu. The method comprises steps as follows: providing a feedbacked implementable drug delivery device (FDDD) with at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; the envelope comprises at least one controllable path for IP's fluid to both inlet and outlet the layer for controlled expulsion of the medication towards the IP by the fluid; further providing the FDDD with a fluid's f low controlling means; and regulating the path by means of the controlling means, thereby controlling the fluid flow.
It is also in the scope of the invention wherein the method as defined above further comprises a step of selecting the fluid flow from a group consisting of IP's f lu id inf low, the medication's outf low or a combination of the same.
It is also in the scope of the invention wherein the method as defined above further comprises a step of communicating the sensing means with the controlling means, and one or more steps ol analyzing one or more members of a group consisting of IP's parameters and targeted region of the body, thereby controlling the f luid flow in a feedbacked manner.
It is also in the scope of the invention wherein the implementable drug delivery device as defined in any of the above further comprising rad iopaque materials, especially metals, barium sulfate or iodine.
It is also in the scope of the invention wherein the implementable drug delivery device, as defined in any of the above, further comprising a net-like layer faci litating the controlled release ol the medication.
It is also in the scope of the invention wherein the implementable drug del ivery device, as defined in any of the above, further comprising a net-like layer facilitating the controlled release of the medication.
It is also in the scope of the invention wherein the implementable drug delivery device, as defined in any of the above, further comprising a pain relieving agents as medications, such as analgesic compositions.
In another embodiment of the invention, a cluster-bomb-like implementable drug delivery system, comprising at least one first envelope containing at least one first medication to be released from the envelope in predefined release profile; wherein the envelope is adapted to be deployed within the intraperitoneal (IP).
Reference is now made to Figs 7 to 12 which further illustrate a ciuster-bomb-like implementable drug delivery system having several envelopes to accommodate a at least one first medication . The envelop may further adapted for loading and releasing at least one first medication and/or at least one second medication respectively. Furthermore, at least one medication may be a controlled release medication.
In another embodiment of the present invention, the envelopes may be in a structure of a chamber, a cell, a shell, a carrier, or a matrix of medication. The envelopes may contain at least one medication in each envelope or severa l medications (i.e., drugs) in one envelope. The envelopes may be configured in a structure selected from a group consisting of a spherical shape (7a-b) , cylindrical shape (8a-b), a ring or tablet-like shape (9a-b, lOa-b) , a rectangu lar shape (8a- b), a tubular shape(lla-b-12a-b) a capsule shape, or any polygon known in the art.
In another embodiment of the present invention, the envelope may further comprise at least one aperture 100 for medications' release. The envelope comprises a distal portion and a proximal portion whilst the proximal portion comprises at least one aperture adapted to be opened to permit medication to eject via the IP to a target region in a patient body. The distal portion may comprise at least one aperture or may additiona lly comprise braking means adapted to bring the cluster bomb-like to a sudden deceleration or acceleration, whereby when the braking means bring the cluster bomb-like to the sudden deceleration or acceleration the at least one first medication is ejected through the proximal portion. The device may further comprise at least one channel 110, 120 having a tube-like, ring-like or/and cylindrical shape for directing at least one medication to the evlepoe's aperture, thereby releasing the medication via the IP route.
It is also in the scope of the invention, wherein at least one of the medication is contained, doped , coated, d issolved, accommodated , im mersed , d iffused, infused, absorbed or otherwise provided in connection with the envelope.
It is also in the scope of the invention, wherein the medication's release is characterized by a profile selected from the group consisting of: a parabo lic release profile, hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
It is also in the scope of the i nvention, wherein the envelop is configured to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly- button, rectum, vagina and any combination thereof.
It is also in the scope of the invention, wherein the envelop is configured to be introduced or implanted via a guiding tool such as catheter.
It is also in the scope of the i nvention ,wherei n the at least one first medication having one profile whilst a second medication having another profile ;the profile is selected from the group consisting of a parabolic release profile, a hyperbolic release, profi le, an exponential release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, an envelope device for delivering at least one first control led release medication form the intraperitoneal (IP) to a target region in a patient body, the at least one first medication is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the envelop; wherein the controlled release is characterized by a profile selected from the group consisting of: a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
In another em bodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed. The system, comprising at least one first enveloping layer and at least one first drug which is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the envelope; wherein the enveloping layer is characterized by a releasing means, and the releasing means are selected from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity; the releasing means provides the system with a drug's release profile selected from, a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitonea l (IP) to a target region located with a patient' body, as defined in any of the above is disclosed. The system further eomp>rising at least one second enveloping layer in which the at least one first drug is contained, carried, doped, coated, d issolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with the second enveloping layer.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region, located with, a patient' body, as defined in any of the above is disclosed. The system further comprising at least one second drug.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed. T he at least one second drugs is contained, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with i the first or second enveloping layers.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed. The system first drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an. exponential
5.1 release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed. The system is configured by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
The drug delivery system of claim 110, wherein the system is configured by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed. The second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, an implantable drug delivery system for delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above is disclosed. The system first and second drug's release profile are different.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body is disclosed. The method comprising steps as follows: providing an implantable drug delivery system with at least one first enveloping layer and at least one first drug; containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer; providing the least one first enveloping layer with an effective a releasing means, by selecting the releasing means from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity; providing, by the releasing means, the system with a drug's release profile, and selecting the profile from a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is disclosed. The method further comprising steps of providing the system with at least one second enveloping layer; and containing, carrying, eloping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one first enveloping layer.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is d isclosed . The method further comprising step of providing the system at least one second drug.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is disclosed. The method further comprising steps as follows: providing the system with at least one second enveloping layer; and containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one first drug with the at least one or second enveloping layer.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is disclosed. The method further comprising steps as follows: providing the system with at least one second drug; and containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing the at least one second drug with the at least one or second enveloping layer.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is disclosed. The method further comprising a step of providing the system's at least one first drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is d isclosed . The method iurther comprising a step of providing the system's at least one first second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
In another embodiment of the invention, a method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, as defined in any of the above, is disclosed. The method further comprising a step of configuring the drug delivery system by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
In another embodiment of the invention, a method of implanting a drug delivery system as defined in any of the above id disclosed. The method comprising step of introducing or implanting the same via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
In another embodiment of the invention, a method of implanting a drug delivery system as defined in any of the above id disclosed. The method step of configuring the system by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
In another embodiment of the invention, a method of implanting a drug delivery system as defined in any of the above id disclosed. The niethod further coniprising a step of configuring first and second drug's release profiles to be different.
Reference in now made to Figs. 1.3-19 which further presents release profiles of the envelope device delivering a controlled release medication via the IP fluid. In accordance with the present invention, Figs. 13 and 17 present a release polynomial profile thereby, the medication release rate (t) decreases for a period of time ,(ii) increases for a period of time , (iii) exhibits a zero order release for a period of time and a combination thereof. The release profile may further include a parabolic release curve, a hyperbolic curve and a combination thereof, relative to time. Furthermore, the release profile thereby, the release of the med ication may be adjusted by the medication controlled release system, or/and by the device configuration having apertures controlling the medication release rate relative to time.
In another em bodiment of the present invention, Fig. 14 further presents a linear release profile thereby, the medication release rate may exhibits a zero order release for a period of time i.e constantly , increase linearly or decrease linearly with time. Fig. 15 presents an exponential release profile thereby, the medication release rate decreases exponentially with time until the medication is exhausted. Fig, 16 presents an exponential release profile thereby, the medication release rate increases exponentially with time as d e envelope and/or the medication moves forward to the target region.
In another embodiment of the present invention. Fig. 18-19 further illustrates a release profile of at least one first medication and a second medication or additional dosage based upon a controlled release system such that Fig. 18 presents an exponential release growth of one first med ication (red) and an exponential release decline (blue) of a second medication. In another embodiment, Fig. 19 presents a polynomial release profile of the first and second medication

Claims

1. An implementable drug delivery device (DL)D) for delivering a medication via the intraperitoneal (IP) to a target region in a patient body, said DDD comprising:
a. at least one envelope with at least first layering surface for contain ing a medication; and
b. at least one first volume confined by said envelope, comprising effective measure of said medication;
wherein said envelope comprises at least one ingress path for IP's fluid to both in let and outlet said layer for expulsion of said medication towards the IP by said fluid.
2. A biocompatible drug delivery device (DDD) for activating and delivering at least one activated medication via the intraperitoneal (IP) route to a target region in a patient body, said DDL) comprising:
a. a hollow container having an external surface and internal surface tor housing an inactive medication, said inactive medication renderable active by contact with the IP fluid;
b. at least one first chamber for loading said medication into said hollow container; and,
c. at least one second chamber for releasing said at least one activated medication into the interior body cavity;
wherein said external surface is provided with a functiona l separation element (FSE) having least one protuberance or/and indentation for distancing said hollow container from interior walls of said body cavity; further wherein said first and second chambers provide an ingress path tor IP fluid to enter said hollow container and an exit path for expulsion of activated medication.
3. A biocompati ble drug delivery device for the delivery of at least one first medication to patient's body cavity via the intraperitoneal (IP) route to at least one target region in a patient body, comprising: an implant for housing and releasing said medication, wherein said implant further comprises at least one chamber for containing said drug; said implant is adapted for introduction into the Peritoneal cavity.
The device according to claims 1 -3, wherein said HSE is an outer coating for surrounding said device adapted as a shielding element; said outer coating is adapted for preventing contact of said device with said body tissue and further preventing increment in medication release.
The device according to claims 1-3, wherein said container has a 3D shape selected from the group consisting of a tubular, a cylinder, a roll, a tube, a cube and combination thereof.
The device according to claims 1-3, wherein said ingress path and exit path are the same path or two separated paths.
The device according to claims 1-3, wherein I P fluid contains an actuation element for activating said medication; said actuation element is selected from the group consisting of I P fluid 's water, IP fluid 's electrolytes, I fluid 's antibodies, IP fluid !s white blood cells, IP fluid 's bio-chemicals and combination thereof.
The device according to claims 1-3, wherein said external surface comprises at least one actuation flap movable by I fluid flow; said flap allows movement of said device in said I P fluid and further promotes said medication delivery.
The device according to claims 1-3, wherein said drug-releasing component is a selective barrier element attached to at least first chamber selected from a group consisting of membrane, a mesh or a net like having at least one pore allowing some particles or chemicals to pass through.
The device according to claims 1-3, wherein said device additionally comprising an anchoring means adapted for securing said device to a body tissue.
The device according to claims 1-3, wherein said medication is selected from a group consisting of a liquot, tablet, cap let, capsule, pill, bolus, particle, micron ized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
The device according to claims 1-3, wherein said medication is in a liquid phase, gas phase or solid phase.
13. The device according to claims 1-3, wherein additionally comprising a medication based upon a controlled release delivery system.
14. The device according to claim 13, wherein said controlled release delivery system is selected from the group consisting of: s!ow release, immediate release, sustained release and combination thereof.
15. The device according to claims 1-3, wherein said device is a biodegradable containing said medication that has been physically trapped, or covalently or ionicaHy immobilized in said biodegradable matrix.
16. The device according to claim 15, wherein said biodegradable is selected from the group consisting of: liposomes, polylactides (polylactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycolide (co-polymers of lactic acicl and glycolic acid) po!yanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin su lfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
17. The device according to cla ims 1-3, wherein said device is a non-absorbable, flexible container having an elongated shape.
18. The device according to claims 1-3. wherein said at least first medication is at a rate of at least approximately 0.1 meg/day over a period of at least approximately one week.
19. The device according to claims 1-3, wherein said device is used tor treating Endometriotic disease.
20. The device according to claims 1-3, wherein said device is semi permeable or permeable to said IP fluid.
21. The device according to claims 1-3, wherein said region is selected from the group consisting of: rectum, large intestine, smal l intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fal lopian tubes, urethra, bladder and, intra-articular cavity and com bination thereof.
22. The device according to claims 1 -3, wherein said medication is transported to the Peritoneal flu id via diffusion, u ltrafiltration, fluid re-absorption and a combination thereof.
23. The device according to claims 1-3, wherein said device is affixed and secured to said body tissue using an applicator via anchoring means.
24. The device according to claims 1-3, wherein said medication is situated at any one of the following locations selected from the group consisting of: a. external surface of the device, b. internal surface of device within at least one of the chambers and, c. within a network of pores of which the device comprises.
25. The device according to claims 1-3, wherein further comprising a monitor unit for measuring physical or/and chemical characteristics within the device; said monitor further adapted to control medication dosage release.
26. A method for administering a medication to a patient, comprising steps of:
a. providing biocompatible drug delivery device for activating and delivering at least one activated medication via the intraperitoneal (IP) route to at least one target region in a patient body comprising:
i. a hol low container having an external surface and internal surface for housing an inactive medication, said inactive medication renderable active by contact with the I P fluid;
ii. at least one first chamber for loading said at least one inactive medication into said hollow container; and,
iii. at least one second chamber for releasing said at least one activated medication into the interior body cavity;
b. introducing said drug delivery device to said patient's body cavity;
wherein said method additionally comprising step of providing said device external surface with a functional separation element (FSE) having least one protruberance or/and indentation for d istancing said hollow container from interior walls of said body cavity said FSE; further wherein said method additionally comprising step of providing said chambers comprising an ingress path for I P fluid to enter said hollow container and an exit path for expulsion of activated medication.
27. The method according to claim 26, wherein additionally comprising step of providing said device having an outer coating surrounding said device as a shielding element; said outer coating is adapted for preventing contact with said body tissue and further preventing increment of the concentration release of said medication
28. The method according to claim 26, wherein said step pf providing said container having a 3 D shape selected from the group consisting of a tubu lar, a cylinder, a rol l, a tube, a cube and a combination thereof,
29. The method according to claim 26, wherein additionally comprising step of providing at least one I P fluid's actuation element for activating said medication; said actuation element is selected from the group consisting of IP fluid 's water, IP fluid 's electrolytes, IP fluid 's antibodies, IP fluid's white blood cells, IP fluid 's bio-chemicals and combination thereof.
30. The method according to claim 26, wherein said method additional comprising step of providing said external surface having at least one protrusion or pin movable by ( P fluid flow; said pin allow movement of said device in said IP fluid and further promoting the medication delivery'.
31. The method according to claim 26, wherein said method additionally comprising step of anchoring and securing said drug delivery device to the body tissue of said patient using anchoring means
32. The method according to claim 26, wherein said releasing said medication is from a first configuration suited for passage of the drug delivery device through said drug-releasing component to a second configuration suited for retaining the drug delivery device with the intraperitoneal (IP) direction route.
33. The method according to claim 26, wherein said medication is selected from a group consisting of a liquot, tablet, cap let, capsule, pill, bolus, particle, micron ized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
4. The method according to claim 26, wherein said method comprising the step ot providing a medication in a liquid phase, gas phase or solid phase,
5. The method according to claim 26, wherein said method comprising the step of providing a medication based upon a controlled release delivery system.
6. The method according to claim 35, wherein said controlled release delivery system is selected from the group consisting ot: slow release, immediate release, sustained release and combination thereof.
7. The method according to claim 26, wherein said device is a biodegradable container containing said medication that has been physically trapped, or covalentiy or ionically immobilized in said biodegradable matrix.
8. The method according to claim 37 , wherein said biodegradable is selected from the group consisting of. liposomes, poiyiacticles (polyiactic acid), polyglycolide (polymer of glycolic acid), poly (lactide co-glycolide (co-polymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvi nyl propylene, polyvinylpyrrolidone and silicone.
9. The method according to claim 26 . wherein said method additionally comprising steps of releasing said at least first medication at a rate ot at least approximately 0.1 meg/day and at least second medication at a rate of at least 0.5 meg/ day over a period of at least approximately three months,
0. The method according to claim 26, wherein said said method additionally comprising steps of providing said device for treating Endometriotic disease.
1. The method according to claim 26 , wherein said said method additiona lly comprising steps of providing said device having a semi permeable characteristics.
2. The method according to claims 26, wherein said method additionally comprising steps of selecting said region from the group consisting of: rectum, large intestine, smaliintestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity and combination thereof.
The method according to claim 26 , wherein said method additionally comprising the step of transporting said medication to the Peritoneal fluid via a) diffusion, b) ultrafi ltration, c) fluid re-absorption and com bination thereof.
The method according to claim 26, wherein additionally comprising step of affixing and securing said device to said body tissue using an applicator via anchoring means.
The method according to claims 26, wherein further comprising step of providing a monitor unit within said device for measuring physical or/ and chemical characteristics within the device; said monitor further adapted to control medication dosage release.
A method for administering a medication to a patient, comprising steps o :
a. providing a biocompatible drug deliver/ device for the delivery of at least one first medication and at least one second medication to a patient's body cavity, said device comprising:
i. at least first chamber and at least one second chamber for loading at least one first medication and said at least one second medication respectively; and,
ii. at least one drug-releasing component located between said chambers, adapted for controlling the release of said first and second medications from said chambers;
b. inserting said drug delivery device to a body cavity of said patient; and c. releasing of said medication via said drug-releasing component into said body cavity of said patient's body;
wherein said step of releasing of said medication via drug-releasing component further comprising step of associating said drug-releasing component with device's external surface and/ or internal surface for controlling the rate of release of said medications; further wherein said method additionally comprising step of delivering said at least first medication and said at least second medication via the intraperitoneal (IP) route to at least one target region in a patient's body ; said at least one medication is actuated when in IP fluid connection.
A method tor administering a medication to a target region within patient's body cavity, comprising steps of:
a. providing a biocompatible drug delivery' device for the delivery of at least a first medication and a second medication to a patient body cavity, said device comprising:
i. at least one first chamber and at least one second chamber for loading at least one first medication and at least one second medication respectively; and,
ii. at least one drug-releasing component located between said chambers adapted for medication release from said chambers; b. inserting said drug delivery device to a body cavity of said patient; and c. releasing of said medication vi drug-releasing component of sa id device into said body cavity of said patient body;
wherein sa id method additionally comprising steps of delivering said at least one first medication to said intraperitoneal (IP) route, second to a first target region and third, to a second target region; further wherein said step b comprising step of functionally associating said drug-releasing component with said device's external surface and/or internal surface for controlling the rate of release of said medications.
A method of manufacturing a biocompatible drug delivery device for the delivery of at least one first medication and at least one second medication to a patient's body cavity, comprising steps of:
a, providing a biocompatible drug delivery device for the delivery of at least a first medication and a second medication to a patient body cavity, said device comprising:
i. at least one first chamber and at least one second chamber for loading at least one first medication and at least one second medication respectively; and,
ii. at least one drug-releasing component located between said chambers adapted for medication release from said chambers; b. connecting at least one drug-releasing component between said at least one first chamber and said at least one second chamber ; said drug-releasing component is for medication release;
wherein additionally comprising steps of associating said drug-releasing component with said at least device external surface and/or internal surface for controlling the rate of release of said at least first medication via the intraperitoneal (IP) route to at least two target regions in a patient's abdominal cavity.
A method for administering a device to a target region within a patient's body cavity, comprising steps of:
a. providing a biocompatible drug delivery device for the delivery of at least one first medication and at least one second medication to the intraperitoneal (IP) route, said device comprising:
i. at least one first chamber and at least one second chamber for loading at least one first medication and at least one second niedication_respectively; and,
is, at least one drug-releasing component located between said chambers adapted for medication release from said chambers; and,
b. providing an applicator adapted for loading said device;
c. placing said drug delivery device in the end of said applicator ;
d . inserting said end of said applicator to a target region within patient's body cavity; e. releasing said device from said end of said applicator whereby said device automatically seats itself within said patient's body cavity; and,
f. withdrawing said applicator from said patient's body cavity;
wherein said method additionally comprising step of delivering said at least one first medication and said at least one second med ication via the intraperitoneal (I P) route to at least two target regions in said patient's body; said at least one medication is actuated when in I P fluid connection.
The method according to claims 46 to 49, wherein said drug-releasing component is selected from the group consisting of: a selective barrier element attached to at least first chamber, selected from the group consisting of membrane, a mesh or net like having at least one pore allowing some particles or chemicals to pass through.
51. The method according to claims 46 to 49, wherein said method additionally comprising step of providing high concentration of said medication when located in said IP route.
52. The method according to claims 46 to 49, wherein said method additionally comprising step of anchoring said drug delivery device to the body tissue of said patient using anchoring means.
53. The method according to claim 52, wherein said method additionally comprising step of attaching anchoring means to at least one edge of said device.
54. The method according to claims 46 to 49, wherein said step of releasing said medication is from a first configuration suited for passage of the drug delivery device through said drug-releasing component to a second configuration suited for retaining the drug delivery- device with the intraperitoneal (IP) direction route.
55. The method according to claims 46 to 49, wherein said said method additionally comprising steps of selecting said medication from a group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates or any combination thereof.
56. The method according to claims 46 to 49, wherein said method additionally comprising step of providing a medication in a liquid phase, gas phase or solid phase.
57. The method according to claims 46 to 49, wherein said method additionally comprising step of providing a medication based upon a controlled release delivery system.
58. The method according to claim 57, wherein said additionally comprising step of selecting said controlled release delivery system from the group consisting of: slow release, immediate release, sustained release and combination thereof.
59. The method according to claims 46 to 59, wherein said step of providing said device having a biodegradable container containing said medication that has been physically trapped, or covaiently or ionicaily immobilized in said biodegradable matrix.
60. The method according to claim 59, wherein said said method additionally comprising steps of selecting said biodegradable from the group consisting of: liposomes, polylactides (polylactic acid), polygiycolide (polymer of glycolic acid), poly (lactide co- glycoiicle (co-polymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, po lyvinyl propylene, polyvinylpyrrolidone silicone and any combination thereof.
61. The method according to claims 46-49, wherein said step of providing said device comprising a container having a 3 D shape selected from the group consisting of a tubular, a cylinder, a roll, a tube, a cube and combination thereof,
62. The method according to claims 46 to 49, wherein said step of providing said device as a non-absorbable, flexible container having an elongated shape.
63. The method according to claim 46 to 49, wherein said method additionally comprising step of releasing said at least first medication at a rate of at least approximately 0.1 meg/ day ana at least second medication at a rate of at least 0.5 meg/day over a period of at least approximately one week.
64. The method according to claims 46 to 49, wherein said device is used for treating Endometriotic disease.
65. The method according to claims 46 to 49, wherein said method additionally comprising steps of selecting said region from the group consisting of: rectum, large intestine, small intesti e, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity and combination thereof.
66. The method according to claims 46 to 49, wherein said method comprising the step of transporting said medication to the Peritoneal fluid via a) diffusion , b) ultrafiltration, c) fluid re-absorption and combination thereof.
67. The method according to claims 46 to 49, wherein additionally comprising step of affixing and securing said device to said body tissue using an applicator via anchoring means.
68. A biocompatible drug delivery device for the delivery of at least one first medication and at least one second medication to patient's body cavity, comprising:
a. at least one first chamber and at least one second chamber for loading said at least one first medication and said at least one second medication respectively; and,
b, at least one drug-releasing component located between said chambers adapted for medication release from said chambers;
wherein said drug-releasing component is associated with said device external surface and/or internal surface for control ling the rate of release of said medications dosage; further wherein said at least one first medication is delivered via the intraperitoneal (I P) route to at least two target regions in a patient body; said at least one medication is actuated when, in IP flu id connection.
69. The device according to claim 68 , wherein said drug-releasing component is a selective barrier element attached to at least first chamber, selected from the group consisting of membrane, a mesh or net li ke having at least one pore allowing some particles or chemicals to pass through, but not other..
70. The device according to claim 68, wherein said device is biodegradable.
71. The device according to claim 68 , wherein further comprising an anchoring means for securing said device to a body tissue.
72. The device according to claim 68 , wherein said medication is selected from a group consisting of aliquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granu le, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates and any combination thereof
73. The device according to claim 68 , wherein said medication is selected from a group consisting of a liquot, tablet, caplet, capsule, pill, bolus, particle, micronized particle, particulate, pellet, core, powder, granule, granulate, small mass, seed, specks, spheres, crystals, beads, agglomerates and any combination thereof.
74. The device according to claim 68, wherein said medication is based upon controlled release delivery system.
75. The device according to claim 68, wherein said controlled release delivery system is selected from the group consisting of: slow release, immediate release, sustained release and combination thereof.
76. The device according to claim 68, wherein said device is a biodegradable container containing said medication that has been physically trapped, covalently or ioniealiy immobilized in a biodegradable matrix.
77. The device according to claim 68, wherein said biodegradable is selected from the group consisting of: liposomes, polylactides, polyglycoiide, poly (lactide co-glycoiicle (copolymers of lactic acid and glycolic acid) polyanhydrides, polypeptides, hyaluronic acid, collagen, c ondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
78. The device accord ing to claim 76, wherein said container is a 3D shape selected from, the group consisting of a tubular, a cylinder, a roll, a tube, a cube and combination thereof.
79. The device according to claim 68, wherein said device is a non-absorbable, flexible container having an elongated shape.
80. The device according to claim 68, wherein said device is semi permeable.
81. The device according to claim 68 , wherein said at least first medication is released at a rate of at least approximately 0.1 meg/day whilst said at least second medication is released at a rate of at least approximately 0.5 meg/day over a period of at least approximately one week.
82. The device according to claim 68, wherein said device is used for treating Endometriotic disease.
The device according to claim 68, wherein said regions are selected from the group consisting of: rectum, large intestine, small intestine, esophagus, stomach, trachea, bronchus, outer ear canal, inner ear canal, nasal canal, air sinuses, vagina, cervix, uterus, fallopian tubes, urethra, bladder and, intra-articular cavity and combination thereof.
The device according to claim 68, wherein said at least first said medication is transported via a) diffusion, b) ultrafiltration, c) fluid re-absorption and combination thereof.
The device according to claim 68, wherein said device is useful for treating endometriosis
The device according to claim 68, wherein said device additionally comprising an applicator for affixing said device to said body tissue via anchoring means.
A kit for the delivery of at least a first medication and a second medication to a patient body cavity, comprising:
a. a biocompatible drug delivery device comprising:
i. at least one first chamber and at least one second chamber for loading said at least one first medication and said at least one second medication respectively? ii. at least one drug-releasing component located between said chambers adapted for medication release from said chambers; and,
b. an applicator adapted for affixing said device to said body tissue;
wherein said drug-releasing component is adapted for controlling the rate of release of said medication from device external surface or/and internal surface; further wherein said at least first medication is delivered via the intraperitoneal (I P) route to at least two target regions in a patient body; said at least one medication is actuated when in IP fluid connection.
The kit according to claim 87, wherein said applicator is adapted for inserting the device into the body of a patient or/and for removing the device from said patient.
The kit according to claim 87, wherein said device further comprising an anchoring means for securing said device to a body tissue.
90. The kit according to claim 87, wherein said applicator is further adapted for reloading said first medication and/or second medication into said device chambers.
91. The kit according to claim 87, wherein said kit is useful for treating endometriosis.
92. An implementable drug delivery pump (DDP) for facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body, said DDP comprising: a. at least one envelope with at least first layering surface for containing a medication; said envelope comprises at least one ingress path for IP's fluid to both inlet and outlet said layer for expulsion of said medication towards the IP by said fluid and further and
b. at least one first volume confined by said envelope, comprising effective measure of said medication; and
c. a medication delivery pump;
wherein said medication delivery pump facilitates the delivery of said medication from said at least one envelope.
93. A method of facilitating the delivery of a medication via the intraperitoneal (IP) to a target region in a patient body, said method comprising:
a. providing an implementable drug delivery pump (DDP) by containing a medication within at least one envelope with at least first layering surface; providing said envelope with at least one ingress path for IP's fluid to both inlet and outlet said layer for expu lsion of said medication towards the IP by said fluid; b. confining by said envelope at least one first volume, comprising effective measure of said medication; and
c. facilitating the delivery of said medication from said at least one envelope by means of a medication delivery pump.
94. An implementable drug delivery device (DDD) for delivering a medication via the intraperitoneal (IP) to a target region in a patient body, said DDD comprising at least one envelope with at least first layering surface confining at least one first volume accomodating at least one first precursor and at least one second precursor; wherein said envelope comprises at least one ingress path for IP's fluid to both inlet and outlet said layer for expu lsion of said first and second precursors towards the IP by said fluid; and further wherein at least one first precursor and at ieast one second precursor forms an effective medication to target said region in a patient body.
95. A method of targeting a region in a patient body, comprising:
a. providing a drug delivery device (DDL)) with at least one envelope, comprising at Ieast first layering surface;
b. confining with in said envelope at least one first precursor and at least one second precursor;
c. providing within said envelope at least one ingress path for IPs fluid to both inlet and outlet said layer for expulsion of said first and second precursors towards the LP by said fluid;
d. forming, by means of said at least one first precursor and at least one second precursor, an effective medication; and
e. targeting said newly formed medication said region in a patient body.
96. A feedback implementable drug delivery device (FDDD) for delivering a time-resolved dose of one or more medications via the intraperitoneal (I P) to a target region in a patient body, said FDDD comprising:
a. at least one envelope with at Ieast one first layering surface confining at least one first volume which accommodates at least one medication; said envelope comprises at least one controllable path for IP's fluid to both inlet and outlet said layer for controlled expulsion of said medication towards the IP by said fluid;
b. a fluid's flow controlling means;
wherein said controlling means regulates said path thereby controlling said fluid flow.
97. The FDDD of claim 96, wherein said fluid flow is selected from a group consisting of IP's fluid inflows said medication's outflow or a combination of the same.
98. The FDDD of claim 96, wherein said device further comprising a sensing means in communication with said controlling means, for analyzing one or more members of a group consisting of I P's parameters and targeted region of the body, thereby controlling said fluid flow in a feedback manner.
99. A method of del ivering a time-resolved dose of one or more medications via the intraperitoneal (IP) to a target region in a patient body, said method comprising: a. providing a feedback irnplementable drug delivery device (FDDD) with at least one envelope with at least one first layering surface confining at least one first volume which accommodates at least one medication; said envelope comprises at least one controllable path for IP's fluid to both inlet and outlet said layer for controlled expu lsion of said medication towards the IP by said fluid;
b. further providing said FDDD with a fluid's flow controlling means; and c. regulating said path by means of said controlling means, thereby control ling said fluid flow.
100. The method of claim 99, wherein said method further comprising a step of selecting said fluid flow from a group consisting of IP's fluid inflow, said medication's outflow or a combination of the same.
101. The method of claim 99, wherein said method further comprising a step of communicating said sensing means with said controlling means, and one or more steps of analyzing one or more members of a group consisting of IP's parameters and targeted region of the body, thereby controlling said fluid flow in a feedback manner.
102. The irnplementable drug delivery? device according to any of claims 1, 2, 3, 68, 87,92, 94, 96 and any of its dependent claims, wherein said device further comprising radiopaque materials, especially metals, barium sulfate or iodine.
103. The irnplementable drug delivery device according to any of claims 1, 2, 3, 68, 87,92, 94, 96 and any of its dependent claims wherein said device further comprising a net-like layer facilitating the controlled release of the medication.
104. The irnplementable drug delivery device according to any of claims 1, 2, 3, 68, 87,92, 94, 96 and any of its dependent claims, wherein said device further comprising a net-like layer facilitating the controlled release of the medication.
105. The irnplementable drug delivery device according to any of claims 1, 2, 3, 68, 87,92, 94, 96 and any of its dependent claims, wherein medication is pain relieving agent, such as analgesic compositions.
6. An implantable drug delivery system tor delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient body, comprising at least one first enveloping layer and at least one first drug which is contained, carried, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with said envelope; wherein said enveloping layer is characterized by a releasing means, and said releasing means are selected from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity; said releasing means provides said system with a drug's release profile selected from a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof,
7. The drug delivery system of claim 106, wherein said system further comprising at least one second enveloping layer in which said at least one first drug is contained, carried, eloped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with said second enveloping layer.
8. The drug deliver/ system of claim 106, wherein said system further comprising at least one second d rug.
9. The drug delivery system of claim. 108, wherein at least one of said second drugs is contained, doped, coated, dissolved, accommodated, immersed, diffused, infused, absorbed or otherwise provided in connection with in said first or second enveloping layers.
0. The drug delivery system of claim 106, wherein said first drug's release profi le is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponential release profile, a linear release profile and any combination thereof.
1. The drug delivery system of claim 106, wherein said system is configured by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
2. The drug delivery system of claim 106, wherein said system is configured by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
3. The drug delivery system of claim 108, wherein said second drug's release profile is selected from a group consisting of a parabolic release profile; hyperbolic release profile, an exponentia l release profi le, a linear release profile and any combination thereof.
4. The drug delivery system of claim 113, wherein said first and second drug's release profile are different.
5. A method of delivering at least one first drug form the intraperitoneal (IP) to a target region located with a patient' body, comprising:
a. providing an implantable drug delivery system with at least one first enveloping layer and at least one first drug;
b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing said at least one first drug with said at least one first enveloping layer;
c. providing said least one first enveloping layer with an effective a releasing means, by selecting said releasing means from physical means, especially porosivity parameters, and chemical means, especially solubility and selectivity;
cl. providing, by said releasing means, said system with a drug's release profile, and e. selecting said profile from a group consisting of a parabolic release profile, a hyperbolic release profile, an exponential release profile, a linear release profi le and any combination thereof.
6. The method of claim 115, further comprising
a. providing said system with at least one second enveloping layer;
b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing said at least one first drug with said at least one first enveloping layer;
7. The method of claim 115, further comprising step of providing said system at least one second drug.
8. The method of claim 115, further comprising steps as follows-,
a. providing said system with at least one second enveloping layer; b. containing, carrying, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing said at least one first drug with said at least one or second enveloping layer.
The method of claim 1 15, further comprising steps as follows:
a. providing said system with at least one second drug;
b. containing, earning, doping, coating, dissolving, accommodating, immersing, diffusing, infusing, absorbing, or otherwise providing said at least one second drug with said at least one or second enveloping layer.
The method of claim 1 15, further comprising a step of providing said system's at least one first drug's release profile is selected from a group consisting of a parabolic release profile: hyperbolic release profi le, an exponential release profile, a linear release profile and any combination thereof.
The method of claim 1 15, further comprising a step of providing said system's at least one first second drug's release profile is selected from a group consisting of a parabolic release profi le; hyperbolic release profile, an exponentia l release profile, a linear release profile and any combination thereof.
The method of cla im 1 15, further comprising a step of configu ring said drug delivery system by means of size of shape to be introduced or implanted via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
A method of implanting a drug delivery system as defined in claim 106 comprising step of introducing or implanting the same via a natural body orifice selected from the group consisting of Douglas pouch, belly-button rectum, vagina and any combination thereof.
A method of implanting a drug delivery system as defined in claim 106 comprising step of configuring said system by means of size or shape to be introduced or implanted via a guiding tool, especially a catheter.
The method of claim 124, further comprising a step of configuring first and second drug's release profiles to be different.
PCT/IL2014/050572 2013-06-25 2014-06-25 Method and device for delivering therapeutic agents to a target region and means thereof WO2014207746A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201361838920P 2013-06-25 2013-06-25
US61/838,920 2013-06-25
US201361847571P 2013-07-18 2013-07-18
US61/847,571 2013-07-18
US201361887394P 2013-10-06 2013-10-06
US61/887,394 2013-10-06

Publications (1)

Publication Number Publication Date
WO2014207746A1 true WO2014207746A1 (en) 2014-12-31

Family

ID=52141182

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2014/050572 WO2014207746A1 (en) 2013-06-25 2014-06-25 Method and device for delivering therapeutic agents to a target region and means thereof

Country Status (1)

Country Link
WO (1) WO2014207746A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022262862A1 (en) * 2021-06-17 2022-12-22 苏州医本生命科技有限公司 Medical particle storage tube, drug loading method, and method for delivering drug-loaded microparticles

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4449983A (en) * 1982-03-22 1984-05-22 Alza Corporation Simultaneous delivery of two drugs from unit delivery device
US7699834B2 (en) * 2005-11-09 2010-04-20 Searete Llc Method and system for control of osmotic pump device

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4449983A (en) * 1982-03-22 1984-05-22 Alza Corporation Simultaneous delivery of two drugs from unit delivery device
US7699834B2 (en) * 2005-11-09 2010-04-20 Searete Llc Method and system for control of osmotic pump device

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022262862A1 (en) * 2021-06-17 2022-12-22 苏州医本生命科技有限公司 Medical particle storage tube, drug loading method, and method for delivering drug-loaded microparticles

Similar Documents

Publication Publication Date Title
US20180339143A1 (en) Implantable device for intraperitoneal drug delivery
JP5265359B2 (en) Intravesical drug delivery device and method
JP7383681B2 (en) Drug delivery system and method for treating bladder cancer with gemcitabine
Ghosh et al. Drug delivery through osmotic systems–an overview
KR101538748B1 (en) Implantable drug delivery device and methods for treatment of the bladder and other body vesicles or lumens
US10406335B2 (en) Drug delivery systems and methods for treatment of prostate
KR102157548B1 (en) Drug delivery systems and methods for treatment of bladder cancer comprising oxaliplatin
KR20150058271A (en) Drug delivery systems and methods for treatment of bladder voiding dysfunction and other lower urinary tract disorders by using trospium
JP2017508749A5 (en)
JP3399527B2 (en) Long-term administration device containing hydrophobic loading drug
WO2014207746A1 (en) Method and device for delivering therapeutic agents to a target region and means thereof
Ali et al. Chronopharmaceutics: a promising drug delivery finding of the last two decades
Das et al. A Systematic Review on Drug Delivery Systems Based on Their Mechanism of Drug Release and Their Applications
Florence et al. Drug delivery and targeting
Kar Osmotically Controlled Drug Delivery Systems: A Review
McConville Implantable drug delivery devices
JPH02185264A (en) Method and device for introducing remedy into bladder or ureter

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14817974

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14817974

Country of ref document: EP

Kind code of ref document: A1