US20100239632A1 - Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue - Google Patents

Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue Download PDF

Info

Publication number
US20100239632A1
US20100239632A1 US12/409,058 US40905809A US2010239632A1 US 20100239632 A1 US20100239632 A1 US 20100239632A1 US 40905809 A US40905809 A US 40905809A US 2010239632 A1 US2010239632 A1 US 2010239632A1
Authority
US
United States
Prior art keywords
drug depot
analgesic
approximately
day
depot
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/409,058
Inventor
Andrew J. Lowenthal Walsh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Companion Spine LLC
Warsaw Orthopedic Inc
Original Assignee
Warsaw Orthopedic Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Warsaw Orthopedic Inc filed Critical Warsaw Orthopedic Inc
Priority to US12/409,058 priority Critical patent/US20100239632A1/en
Assigned to MEDTRONIC, INC. reassignment MEDTRONIC, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WALSH, ANDREW J. LOWENTHAL
Priority to PCT/US2010/028140 priority patent/WO2010111178A2/en
Priority to EP10756666A priority patent/EP2410998A4/en
Publication of US20100239632A1 publication Critical patent/US20100239632A1/en
Priority to US15/280,381 priority patent/US20170014337A1/en
Priority to US16/223,571 priority patent/US10653619B2/en
Assigned to COMPANION SPINE, LLC reassignment COMPANION SPINE, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MEDTRONIC INC.
Assigned to COMPANION SPINE, LLC reassignment COMPANION SPINE, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MEDTRONIC INC.
Abandoned legal-status Critical Current

Links

Images

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
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F13/00Bandages or dressings; Absorbent pads
    • A61F13/15Absorbent pads, e.g. sanitary towels, swabs or tampons for external or internal application to the body; Supporting or fastening means therefor; Tampon applicators
    • A61F13/20Tampons, e.g. catamenial tampons; Accessories therefor
    • A61F13/2002Tampons, e.g. catamenial tampons; Accessories therefor characterised by the use
    • A61F13/2005Tampons, e.g. catamenial tampons; Accessories therefor characterised by the use specially adapted for the nose cavity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F13/00Bandages or dressings; Absorbent pads
    • A61F13/15Absorbent pads, e.g. sanitary towels, swabs or tampons for external or internal application to the body; Supporting or fastening means therefor; Tampon applicators
    • A61F13/84Accessories, not otherwise provided for, for absorbent pads
    • A61F13/8405Additives, e.g. for odour, disinfectant or pH control
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41681,3-Diazoles having a nitrogen attached in position 2, e.g. clonidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4468Non condensed piperidines, e.g. piperocaine having a nitrogen directly attached in position 4, e.g. clebopride, fentanyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/451Non condensed piperidines, e.g. piperocaine having a carbocyclic group directly attached to the heterocyclic ring, e.g. glutethimide, meperidine, loperamide, phencyclidine, piminodine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4535Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom, e.g. pizotifen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/655Azo (—N=N—), diazo (=N2), azoxy (>N—O—N< or N(=O)—N<), azido (—N3) or diazoamino (—N=N—N<) compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1282Devices used in vivo and carrying the radioactive therapeutic or diagnostic agent, therapeutic or in vivo diagnostic kits, stents
    • 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
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7007Drug-containing films, membranes or sheets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • Pain and inflammation relief is of prime importance to anyone treating patients undergoing surgery. Proper pain and inflammation relief imparts significant physiological and psychological benefits to the patient. Not only does effective pain and inflammation relief mean a smoother more pleasant postoperative course (e.g., mood, sleep, quality of life, etc.) with earlier discharge from medical/surgical/outpatient facilities, but it may also reduce the onset of chronic pain and inflammation syndromes (e.g., fibromyalgia, myalgia, etc.).
  • Pain serves a biological function. It often signals the presence of damage or disease within the body and is often accompanied by inflammation (redness, swelling, and/or burning). In the case of postoperative pain and inflammation it may be a result of the surgery, or other treatments such as, for example, management of acute pain following burns or non-surgical trauma.
  • the goal for postoperative pain and inflammation management is to reduce or eliminate pain and inflammation discomfort with medication that cause minimum or no side effects.
  • Prolonged pain and inflammation can reduce physical activity and lead to venous stasis and an increased risk of deep vein thrombosis and consequently pulmonary embolism.
  • there can be widespread effects on gut and urinary tract motility which may lead in turn to postoperative ileus, nausea, vomiting and urinary retention. These problems are unpleasant for the patient and may prolong hospital stay.
  • Most patients who experience moderate to severe post-operative pain and inflammation often require pain and inflammation control at least in the first 3 days after trauma or surgery.
  • nasal and sinus cavities One area that is ripe for pain and/or inflammation due to trauma or surgery is the nasal and sinus cavities. Physicians are frequently called upon to treat nasal and sinus cavities as a result of tissue desiccation, trauma, infection, or other nasal and sinus diseases. Another area that is also affected by pain and inflammation is cardiac tissue. For example, during a myocardial infarction, commonly known as a heart attack, the blood supply to part of the heart is interrupted. The resulting ischemia (restriction in blood supply) and oxygen shortage, if left untreated for a sufficient period, can cause damage and/or death (infarction) of heart muscle tissue resulting in pain and inflammation.
  • myocardial infarction commonly known as a heart attack
  • analgesics and/or anti-inflammatory formulations although effective for short term relief of pain and/or inflammation, require frequent single dose administration every 4 to 12 hours on an as needed basis.
  • These single dose analgesics and/or anti-inflammatory formulations are inconvenient and may interfere with the patient's postoperative inpatient and/or outpatient daytime activities and nighttime sleep and recovery.
  • New analgesics and/or anti-inflammatory compositions and methods are needed to treat or reduce postoperative pain and/or inflammation at or near cardiac tissue or within the nasal or sinus cavity.
  • New analgesics and/or anti-inflammatory compositions and methods that reliably provide long acting analgesic and anti-inflammatory effects over periods of 3 to 10 days are needed.
  • New compositions and methods are provided that effectively prevent, treat or reduce postoperative pain or inflammation in areas at or near cardiac tissue or within the nasal or sinus cavity.
  • analgesic and/or anti-inflammatory compositions and methods are provided that have long acting analgesic and/or anti-inflammatory effects over periods of 3 to 10 days in a single drug depot or multiple drug depots.
  • New drug depot films and methods are provided, which can easily allow accurate and precise implantation of a drug depot containing analgesic and/or anti-inflammatory compositions.
  • analgesic and/or anti-inflammatory drug depot compositions and methods can now be easily delivered to the target tissue site (e.g., nasal, sinus and/or cardiac tissue, surgical wound or incision, etc.) and provide pain relief for 3 to 10 days. In this way, accurate and precise implantation of the drug depot can be accomplished.
  • the drug depot is in the form of a film and can be locally delivered to the nasal, sinus and/or cardiac tissue by packing the drug depot at the target tissue site.
  • an implantable drug depot useful for reducing, preventing or treating pain and/or inflammation in a patient in need of such treatment, the implantable drug depot being in the form of a biodegradable film and comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent, the depot being implantable at or near a cardiac tissue or within the nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is capable of releasing an effective amount of the analgesic and/or an anti-inflammatory agent over a period of at least one day.
  • a method of treating or preventing pain and inflammation in a patient in need of such treatment comprising administering one or more biodegradable drug depots comprising a therapeutically effective amount of an analgesic and an anti-inflammatory agent at or near a cardiac tissue or within the nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is in the form of a biodegradable film or strip that releases an effective amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of at least 1 day.
  • a method for reducing pain and inflammation in a patient in need of such treatment comprising delivering one or more biodegradable drug depots in the form of a biodegradable film comprising a therapeutically effective amount of an analgesic and an anti-inflammatory agent or pharmaceutically acceptable salts thereof at or near a cardiac tissue or within the nasal or sinus cavity of the patient, wherein the drug depot releases an effective amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of at least 1 day.
  • the therapeutic agent may for example, be part of a drug depot.
  • the drug depot may: (i) consist of the analgesic and/or an anti-inflammatory agent and the biodegradable polymer(s); or (ii) consist essentially of the analgesic and/or an anti-inflammatory agent; or (iii) comprise the analgesic and/or an anti-inflammatory agent and one or more other active ingredients, surfactants, excipients or other ingredients or combinations thereof.
  • these other compounds or combinations thereof comprise less than 20 wt. %, less than 19 wt. %, less than 18 wt. %, less than 17 wt.
  • FIG. 1A illustrates a magnified top view of one embodiment of the implantable drug depot in the form of a film or strip that has the analgesic and/or an anti-inflammatory agent disposed on or in the film or strip.
  • FIG. 1B illustrates a magnified top view of one embodiment of the implantable drug depot in the form of multiple films or strips that have the analgesic and/or an anti-inflammatory agent disposed on or in the films or strips.
  • the films or strips are shown stacked together and can be used as packing material to pack at or near a cardiac tissue or within the nasal or sinus cavity.
  • FIG. 2 illustrates a magnified side view of one embodiment of the implantable drug depot in the form of a sponge that has the analgesic and/or an anti-inflammatory agent disposed on or in the sponge.
  • FIG. 3 illustrates a partial, side sectional view of a human head illustrating the parts of the nasal cavity with the drug depot in the form of a film or strip packing administered thereto.
  • FIG. 4 illustrates a partial, front sectional view of a human head illustrating portions of a nasal cavity and sinus cavity with the drug depot in the form of a film or strip administered to the sinus frontilis.
  • FIG. 5 schematically shows tissues and vessels of the heart where the drug depot in the form of multiple films, or strips, or cardiac patch that can be administered thereto.
  • a drug depot includes one, two, three or more drug depots.
  • New compositions and methods are provided that effectively prevent, treat or reduce postoperative pain or inflammation in areas at or near cardiac tissue or within the nasal or sinus cavity.
  • analgesic and/or anti-inflammatory compositions and methods are provided that have long acting analgesic and/or anti-inflammatory effects over periods of 3 to 10 days in a single drug depot or multiple drug depots.
  • New drug depot films and methods are provided, which can easily allow accurate and precise implantation of a drug depot containing analgesic and/or anti-inflammatory compositions.
  • analgesic and/or anti-inflammatory drug depot compositions and methods can now be easily delivered to the target tissue site (e.g., nasal, sinus and/or cardiac tissue, surgical wound or incision, etc.) and provide pain relief for 3 to 10 days. In this way, accurate and precise implantation of the drug depot can be accomplished.
  • the drug depot is in the form of a film and can be locally delivered to the nasal, sinus and/or cardiac tissue by packing the drug depot at the target tissue site.
  • an implantable drug depot useful for reducing, preventing or treating pain and/or inflammation in a patient in need of such treatment, the implantable drug depot being in the form of a biodegradable film and comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent, the depot being implantable at or near a cardiac tissue or within the nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is capable of releasing an effective amount of the analgesic and/or an anti-inflammatory agent over a period of at least one day.
  • a drug depot includes one, two, three or more drug depots.
  • Analgesic refers to an agent or compound that can reduce, relieve or eliminate pain.
  • analgesic agents include but are not limited to acetaminophen, a local anesthetic, such as for example, lidocaine, bupivacaine, ropivacaine, opioid analgesics such as buprenorphine, butorphanol, dextromoramide, dezocine, dextropropoxyphene, diamorphone, fentanyl, alfentanil, sulfentanil, hydrocodone, hydromorphone, ketobemidone, levomethadyl, levorphanol, meperidine, methadone, morphine, nalbuphine, opium, oxycodone, papavereturn, pentazocine, pethidine, phenoperidine, piritramide, dextropropoxyphene, remifentanil, sulfentanil, tilidine, tramadol
  • anti-inflammatory agent refers to an agent or compound that has anti-inflammatory effects. These agents may remedy pain by reducing inflammation.
  • anti-inflammatory agents include, but are not limited to, a statin, sulindac, sulfasalazine, naroxyn, diclofenac, indomethacin, ibuprofen, flurbiprofen, ketoprofen, aclofenac, aloxiprin, aproxen, aspirin, diflunisal, fenoprofen, mefenamic acid, naproxen, phenylbutazone, piroxicam, meloxicam, salicylamide, salicylic acid, desoxysulindac, tenoxicam, ketoralac, flufenisal, salsalate, triethanolamine salicylate, aminopyrine, antipyrine, oxyphenbutazone, apazone, cintazone, flufenamic acid, clonixeril, clon
  • Anti-inflammatory agents also include other compounds such as steroids, such as for example, fluocinolone, cortisol, cortisone, hydrocortisone, fludrocortisone, prednisone, prednisolone, methylprednisolone, triamcinolone, betamethasone, dexamethasone, beclomethasone, fluticasone interleukin-1 receptor antagonists, thalidomide (a TNF- ⁇ release inhibitor), thalidomide analogues (which reduce TNF- ⁇ production by macrophages), bone morphogenetic protein (BMP) type 2 or BMP-4 (inhibitors of caspase 8, a TNF- ⁇ activator), quinapril (an inhibitor of angiotensin II, which upregulates TNF- ⁇ ), interferons such as IL-11 (which modulate TNF- ⁇ receptor expression), and aurin-tricarboxylic acid (which inhibits TNF- ⁇ ), guanidinoethyld
  • anti-inflammatory agents include, for example, naproxen; diclofenac; celecoxib; sulindac; diflunisal; piroxicam; indomethacin; etodolac; meloxicam; ibuprofen; ketoprofen; r-flurbiprofen; mefenamic; nabumetone; tolmetin, and sodium salts of each of the foregoing; ketorolac bromethamine; ketorolac tromethamine; ketorolac acid; choline magnesium trisalicylate; rofecoxib; valdecoxib; lumiracoxib; etoricoxib; aspirin; salicylic acid and its sodium salt; salicylate esters of alpha, beta, gamma-tocopherols and tocotrienols (and all their d, 1, and racemic isomers); methyl, ethyl, propyl, isopropyl, n-buty
  • An anti-inflammatory agent can be a steroid.
  • exemplary steroids include, for example, 21-acetoxypregnenolone, alclometasone, algestone, amcinonide, beclomethasone, betamethasone, budesonide, chloroprednisone, clobetasol, clobetasone, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacort, desonide, desoximetasone, dexamethasone, dexamethasone 21-acetate, dexamethasone 21-phosphate di-Na salt, diflorasone, diflucortolone, difluprednate, enoxolone, fluazacort, fluclorinide, flumethasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocor
  • Examples of a useful statin for treatment of pain and/or inflammation include, but is not limited to, atorvastatin, simvastatin, pravastatin, cerivastatin, mevastatin (see U.S. Pat. No. 3,883,140, the entire disclosure is herein incorporated by reference), velostatin (also called synvinolin; see U.S. Pat. Nos. 4,448,784 and 4,450,171 these entire disclosures are herein incorporated by reference), fluvastatin, lovastatin, rosuvastatin and fluindostatin (Sandoz XU-62-320), dalvastatin (EP Appln. Publn. No.
  • statin may comprise mixtures of (+)R and ( ⁇ )—S enantiomers of the statin.
  • the statin may comprise a 1:1 racemic mixture of the statin.
  • Anti-inflammatory agents also include those with anti-inflammatory properties, such as, for example, amitriptyline, carbamazepine, gabapentin, pregabalin, clonidine, or a combination thereof.
  • salts of alkali metals such as magnesium, calcium, sodium, potassium and ammonium
  • salts of mineral acids such as hydrochloric, hydriodic, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acids
  • salts of organic acids such as tartaric, acetic, citric, malic, benzoic, glycollic, gluconic, gulonic, succinic, arylsulfonic, e.g., p-toluenesulfonic acids, or the like.
  • analgesic agent when referring to an analgesic agent, unless otherwise specified or apparent from context, it is understood that the inventor is also referring to pharmaceutically acceptable salts including stereoisomers.
  • Pharmaceutically acceptable salts include those salt-forming acids and bases that do not substantially increase the toxicity of the compound.
  • salts of alkali metals such as magnesium, calcium, sodium, potassium and ammonium
  • salts of mineral acids such as hydrochloric, hydriodic, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acids
  • salts of organic acids such as tartaric, acetic, citric, malic, benzoic, glycollic, gluconic, gulonic, succinic, arylsulfonic, e.g., p-toluenesulfonic acids, or the like.
  • a “drug depot” is the composition in which at least one anti-inflammatory agent and at least one analgesic agent or the pharmaceutically acceptable salts of either or both are administered to the body.
  • a drug depot may comprise a physical structure to facilitate implantation and retention in a desired site (e.g., nasal cavity, sinus cavity, cardiac site of the patient, particularly at or near a site of surgery, or other site of inflammation, etc.).
  • the drug depot also comprises the drug itself.
  • drug as used herein is generally meant to refer to any substance that alters the physiology of a patient.
  • drug may be used interchangeably herein with the terms “therapeutic agent,” “therapeutically effective amount,” and “active pharmaceutical ingredient” or “API.” It will be understood that unless otherwise specified a “drug” formulation may include more than one therapeutic agent, wherein exemplary combinations of therapeutic agents include a combination of two or more drugs.
  • the drug provides a concentration gradient of the therapeutic agent for delivery to the site.
  • the drug depot provides an optimal drug concentration gradient of the therapeutic agent at a distance of up to about 0.1 cm to about 5 cm from the implant site (e.g., within the nasal and/or sinus cavity or at or near cardiac tissue), and comprises at least one anti-inflammatory agent or its pharmaceutically acceptable salt and/or at least one analgesic agent or its pharmaceutically acceptable salt.
  • a “depot” includes but is not limited to capsules, microspheres, microparticles, microcapsules, microfibers particles, nanospheres, nanoparticles, coating, matrices, wafers, film, strip, ribbon, sponge, patch, pills, pellets, emulsions, liposomes, micelles, gels, or other pharmaceutical delivery compositions or a combination thereof.
  • the drug depot may comprise a pump that holds and administers the pharmaceutical (e.g., anti-inflammatory and/or analgesic).
  • the drug depot has pores that allow release of the drug from the depot. The drug depot will allow fluid in the depot to displace the drug. However, cell infiltration into the depot will be prevented by the size of the pores of the depot.
  • the depot should not function as a tissue scaffold and allow tissue growth. Rather, the drug depot will solely be utilized for drug delivery.
  • the pores in the drug depot will be less than 250 to 500 microns. This pore size will prevent cells from infiltrating the drug depot and laying down scaffolding cells.
  • drug will elute from the drug depot as fluid enters the drug depot, but cells will be prevented from entering.
  • the drug will elute out from the drug depot by the action of enzymes, by hydrolytic action and/or by other similar mechanisms in the human body.
  • Suitable drug depots for use in the present application are described in U.S. Provisional Application No. 61/046,246 filed Apr. 18, 2008, U.S. Provisional Application No. 61/046,218 filed Apr. 18, 2008, U.S. Provisional Application No. 61/046,218 filed Apr. 18, 2008, U.S. Provisional Application No. 61/046,201 filed Apr. 18, 2008, U.S. Ser. No. 12/105,864 filed Apr. 18, 2008 and U.S. Ser. No. 12/105,375 filed Apr. 18, 2008.
  • the entire disclosure of these applications is herein incorporated by reference into the present application.
  • Suitable materials for the depot are ideally pharmaceutically acceptable biodegradable and/or any bioabsorbable materials that are preferably FDA approved or GRAS materials. These materials can be polymeric or non-polymeric, as well as synthetic or naturally occurring, or a combination thereof. In various embodiments, the drug depot may not be biodegradable or comprise material that is not biodegradable.
  • Non-biodegradable polymers include, but are not limited to, various cellulose derivatives (carboxymethyl cellulose, cellulose acetate, cellulose acetate propionate, ethyl cellulose, hydroxypropyl methyl cellulose, hydroxyalkyl methyl celluloses, and alkyl celluloses), silicon and silicon-based polymers (such as polydimethylsiloxane), polyethylene-co-(vinyl acetate), poloxamer, polyvinylpyrrolidone, poloxamine, polypropylene, polyamide, polyacetal, polyester, poly ethylene-chlorotrifluoroethylene, polytetrafluoroethylene (PTFE or “TeflonTM”), styrene butadiene rubber, polyethylene, polypropylene, polyphenylene oxide-polystyrene, poly- ⁇ -chloro-p-xylene, polymethylpentene, polysulfone, non-degradable ethylene-vinyl a
  • the drug depot may comprise non-resorbable polymers as well.
  • These non-resorbable polymers can include, but are not limited to, delrin, polyurethane, copolymers of silicone and polyurethane, polyolefins (such as polyisobutylene and polyisoprene), acrylamides (such as polyacrylic acid and poly(acrylonitrile-acrylic acid)), neoprene, nitrile, acrylates (such as polyacrylates, poly(2-hydroxy ethyl methacrylate), methyl methacrylate, 2-hydroxyethyl methacrylate, and copolymers of acrylates with N-vinyl pyrrolidone), N-vinyl lactams, polyacrylonitrile, glucomannan gel, vulcanized rubber and combinations thereof.
  • polyurethanes include thermoplastic polyurethanes, aliphatic polyurethanes, segmented polyurethanes, hydrophilic polyurethanes, polyether-urethane, polycarbonate-urethane and silicone polyether-urethane.
  • the non-degradable drug depots may need to be removed.
  • a “therapeutically effective amount” or “effective amount” is such that when administered, the drug results in alteration of the biological activity, such as, for example, inhibition of inflammation, reduction or alleviation of pain, improvement in the condition through the reduction in edema etc.
  • the dosage administered to a patient can unless otherwise specified or apparent from context be as single or multiple doses depending upon a variety of factors, including the drug's administered pharmacokinetic properties, the route of administration, patient conditions and characteristics (sex, age, body weight, health, size, etc.), extent of symptoms, concurrent treatments, frequency of treatment and the effect desired.
  • therapeutically effective doses may be less than doses administered by other routes (oral, topical, etc.).
  • the drug dose delivered from the drug depot may be, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or 99.9% less than the oral dosage or injectable dose.
  • systemic side effects such as for example, liver transaminase elevations, hepatitis, liver failure, myopathy, constipation, etc. may be reduced or eliminated.
  • the depot is designed for immediate release. In other embodiments the drug depot is designed for sustained release. In other embodiments, the drug depot comprises one or more immediate release surfaces and one or more sustain release surfaces.
  • sustained release or “sustain release” (also referred to as extended release or controlled release) are used herein to refer to one or more therapeutic agent(s) that is introduced into the body of a human or other mammal and continuously or continually releases a stream of one or more therapeutic agents over a predetermined time period and at a therapeutic level sufficient to achieve a desired therapeutic effect throughout the predetermined time period (e.g., certain dose/per day).
  • Reference to a continuous or continual release stream is intended to encompass release that occurs as the result of biodegradation in vivo of the drug depot, or a matrix or component thereof, or as the result of metabolic transformation or dissolution of the therapeutic agent(s) or conjugates of therapeutic agent(s).
  • sustained release formulations may, by way of example, be created as films, slabs, strip, fibers, sponges, or gels.
  • microparticles, microspheres, microcapsules, spheroids, shaped derivatives or pastes are disposed in the film or sponge.
  • the formulations may be in a form that is suitable for suspension in isotonic saline, physiological buffer or other solution acceptable for injection into a patient.
  • formulations may be used in conjunction with any implantable, insertable or injectable system that a person of ordinary skill would appreciate as useful in connection with embodiments herein including but not limited to parenteral formulations, microspheres, microcapsules, gels, pastes, implantable rods, pellets, films, strips, plates or fibers, etc.
  • immediate release is used herein to refer to one or more therapeutic agent(s) that is introduced into the body and that is allowed to dissolve in or become absorbed at the location to which it is administered, with no intention of delaying or prolonging the dissolution or absorption of the drug.
  • Immediate release refers to the release of drug within a short time period following administration, e.g., generally within a few minutes to about 1 hour.
  • mammal refers to organisms from the taxonomy class “mammalian,” including but not limited to humans, other primates such as chimpanzees, apes, orangutans and monkeys, rats, mice, cats, dogs, cows, horses, etc. In various embodiments, the mammal is a human patient.
  • release rate profile refers to the percentage of active ingredient that is released over fixed units of time, e.g., mcg/hr, mcg/day, mg/hr, mg/day, 10% per day for ten days, etc.
  • a release rate profile may be but need not be linear.
  • the drug depot may be in a film or strip or patch form that releases at least one analgesic agent and at least one anti-inflammatory agent over a period of time.
  • Treating or treatment of a disease or condition refers to executing a protocol, which may include administering one or more drugs to a patient (human, normal or otherwise, or other mammal), in an effort to alleviate signs or symptoms of the disease. Alleviation can occur prior to signs or symptoms of the disease or condition appearing, as well as after their appearance.
  • “treating” or “treatment” includes “preventing” or “prevention” of disease or undesirable condition (e.g., pain and/or inflammation).
  • “treating” or “treatment” does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols that have only a marginal effect on the patient.
  • “Reducing pain and/or inflammation” includes a decrease in pain and/or inflammation and does not require complete alleviation of pain and/or inflammation signs or symptoms, and does not require a cure. In various embodiments, reducing pain and/or inflammation includes even a marginal decrease in pain and/or inflammation.
  • the administration of the effective dosages of at least one analgesic agent and at least one anti-inflammatory agent may be used to prevent, treat or relieve the symptoms of pain and/or inflammation for different diseases or conditions.
  • These diseases/conditions may comprise chronic inflammatory diseases, including, but not limited to sinusitis, (acute and chronic), rhinitis, nasal and or sinus infection, nasal and/or sinus surgery, epitaxis (nose bleeds), sinus bleeding, nasal and/or sinus obstruction, nasal and/or sinus polyps, nasal and/or sinus cancer, nasal and/or sinus trauma.
  • the drug depot may be used to treat, prevent or reduce diseases/conditions, such as cardiovascular disease.
  • Cardiovascular disease (CVD) is a general term used to classify numerous conditions that affect the heart, heart valves, blood, and vasculature of the body.
  • Cardiovascular diseases include coronary artery disease, angina pectoris, myocardial infarction, atherosclerosis, congestive heart failure, hypertension, cerebrovascular disease, stroke, transient ischemic attacks, cardiomyopathy, arrhythmias, aortic stenosis, or aneurysm.
  • “Localized” delivery includes delivery where one or more drugs are deposited at or near or within a tissue, for example, within the nasal or sinus cavities or cardiac tissue or in close proximity (e.g., within about 5 cm, or preferably within 0.1 cm) thereto.
  • a “targeted delivery system” provides delivery of one or more drugs depots, gels or depot dispersed in the gel having a quantity of therapeutic agent that can be deposited at or near the target site (e.g., nasal or sinus cavity or cardiac tissue) as needed for treatment of pain, inflammation or other disease or condition.
  • biodegradable includes that all or parts of the drug depot will degrade over time by the action of enzymes, by hydrolytic action and/or by other similar mechanisms in the human body.
  • biodegradable includes that the depot (e.g., strip, film, sheet, etc.) can break down or degrade within the body to non-toxic components after or while a therapeutic agent has been or is being released.
  • bioerodible it is meant that the depot will erode or degrade over time due, at least in part, to contact with substances found in the surrounding tissue, fluids or by cellular action.
  • bioabsorbable it is meant that the depot will be broken down and absorbed within the human body, for example, by a cell or tissue.
  • Biocompatible means that the depot will not cause substantial tissue irritation or necrosis at the target tissue site.
  • pain management medication includes one or more therapeutic agents that are administered to prevent, alleviate or remove pain entirely.
  • therapeutic agents include anti-inflammatory agents, muscle relaxants, analgesics, anesthetics, narcotics, and so forth, and combinations thereof.
  • the depot e.g., strip, film, patch, sponge
  • the depot can be designed to cause an initial burst dose of therapeutic agent within the first 24 hours, 2 days, 3 days, 4 days, or 5 days after implantation.
  • “Initial burst” or “burst effect” or “bolus dose” refer to the release of therapeutic agent from the depot during the first 24 hours, 2 days, 3 days, 4 days, or 5 days after the depot comes in contact with an aqueous fluid (e.g., synovial fluid, cerebral spinal fluid, etc.).
  • This burst effect is particularly beneficial for the analgesic, while in various embodiments, for the anti-inflammatory agent a more linear release of a longer duration may be desired.
  • the “burst effect” is believed to be due to the increased release of therapeutic agent from the depot.
  • the depot e.g., gel
  • the depot is designed to avoid this initial burst effect.
  • the drug depot comprising at least one analgesic agent or its pharmaceutically acceptable salt and/or at least one anti-inflammatory agent or its pharmaceutically acceptable salt may be co-administered with a muscle relaxant.
  • Co-administration may involve administering at the same time in separate drug depots or formulating together in the same drug depot.
  • Exemplary muscle relaxants include by way of example and not limitation, alcuronium chloride, atracurium bescylate, baclofen, carboxonium, carisoprodol, chlorphenesin carbamate, chlorzoxazone, cyclobenzaprine, dantrolene, decamethonium bromide, camdinium, gallamine triethiodide, hexafluorenium, meladrazine, mephensin, metaxalone, methocarbamol, metocurine iodide, pancuronium, pridinol mesylate, styramate, suxamethonium, suxethonium, thiocolchicoside, tizanidine, tolperisone, tubocuarine, vecuronium, or combinations thereof.
  • the drug depot may also comprise other therapeutic agents or active ingredients in addition to the at least one analgesic agent or its pharmaceutically acceptable salt and/or at least one anti-inflammatory agent or its pharmaceutically acceptable salt.
  • additional therapeutic agents include, but are not limited to, integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, CTLA4-Ig agonists/antagonists (BMS-188667), CD40 ligand antagonists, Humanized anti-IL-6 mAb (MRA, Tocilizumab, Chugai), HMGB-1 mAb (Critical Therapeutics Inc.), anti-IL2R antibodies (daclizumab, basilicimab), ABX (anti IL-8 antibodies), recombinant human IL-10, or HuMax IL-15 (anti-IL 15 antibodies).
  • IL-1 inhibitors such as Kineret® (anakinra) which is a recombinant, non-glycosylated form of the human interleukin-1 receptor antagonist (IL-1Ra), or AMG 108, which is a monoclonal antibody that blocks the action of IL-1.
  • Therapeutic agents also include excitatory amino acids such as glutamate and aspartate, antagonists or inhibitors of glutamate binding to NMDA receptors, AMPA receptors, and/or kainate receptors. It is contemplated that where desirable a pegylated form of the above may be used.
  • examples of other therapeutic agents include NF kappa B inhibitors such as glucocorticoids, antioxidants, such as dilhiocarbamate.
  • additional therapeutic agents suitable for use include, but are not limited to, an anabolic growth factor or anti-catabolic growth factor, analgesic agent, or an osteoinductive growth factor or a combination thereof.
  • Suitable anabolic growth or anti-catabolic growth factors include, but are not limited to, a bone morphogenetic protein, a growth differentiation factor, a LIM mineralization protein, CDMP or progenitor cells or a combination thereof.
  • Suitable analgesic agents include, but are not limited to, acetaminophen, bupivacaine, opioid analgesics such as amitriptyline, carbamazepine, gabapentin, pregabalin, clonidine, opioid analgesics or a combination thereof.
  • Opioid analgesics include, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levorphanol, levophenacylmorphan, lofentanil, me
  • the release of each compound may be for at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or at least fifteen days, or longer.
  • the therapeutic agent also includes its pharmaceutically acceptable salt.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds (e.g., esters or amines) wherein the parent compound may be modified by making acidic or basic salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, or nitric acids; or the salts prepared from organic acids such as acetic, fuoric, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic acid.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, or nitric acids
  • organic acids such as acetic, fuoric, propionic, succinic, glycolic, stearic,
  • Pharmaceutically acceptable also includes the racemic mixtures ((+)—R and ( ⁇ )—S enantiomers) or each of the dextro and levo isomers of the therapeutic agent individually.
  • the therapeutic agent may be in the free acid or base form or be pegylated for long acting activity.
  • the anti-inflammatory agent in the drug depot comprises sulfasalazine.
  • Sulfasalazine is also known as 6-oxo-3-((4-(pyridin-2-ylsulfamoyl)phenyl) hydrazinylidene]cyclohexa-1,4-diene-1-carboxylic acid.
  • Sulfasalazine or a pharmaceutically acceptable salt thereof is available from various pharmaceutical manufacturers.
  • the dosage of sulfasalazine is from approximately 0.005 ⁇ g/day to approximately 3000 mg/day.
  • Additional dosages of sulfasalazine include from approximately 0.005 ⁇ g/day to approximately 2000 mg/day; approximately 0.005 ⁇ g/day to approximately 1000 mg/day; approximately 0.005 ⁇ g/day to approximately 100 mg/day; approximately 0.005 ⁇ g/day to approximately 1 mg/day; approximately 0.005 ⁇ g/day to approximately 80 ⁇ g/day; approximately 0.01 to approximately 70 ⁇ g/day; approximately 0.01 to approximately 65 ⁇ g/day; approximately 0.01 to approximately 60 ⁇ g/day; approximately 0.01 to approximately 55 ⁇ g/day; approximately 0.01 to approximately 50 ⁇ g/day; approximately 0.01 to approximately 45 ⁇ g/day; approximately 0.01 to approximately 40 ⁇ g/day; approximately 0.025 to approximately 35 ⁇ g/day; approximately 0.025 to approximately 30 ⁇ g/day; approximately 0.025 to approximately 25 ⁇ g/day; approximately 0.025 to approximately 20 ⁇ g/day; and approximately 0.025 to approximately 15 ⁇ g/day.
  • the anti-inflammatory agent in the drug depot comprises sulindac.
  • Sulindac also known as 2-[6-fluoro-2-methyl-3-[(4-methylsulfinylphenyl)-methylidene]inden-1-yl]-acetic acid may be represented by the formula C 20 H 17 FO 3 S.
  • Sulindac or a pharmaceutically acceptable salt thereof is available from various pharmaceutical manufacturers.
  • the dosage of sulindac may be from approximately 0.001 ⁇ g/day to approximately 400 mg/day. Additional dosages of sulindac include from approximately 0.001 ⁇ g/day to approximately 200 mg/day; approximately 0.001 ⁇ g/day to approximately 100 mg/day; approximately 0.001 ⁇ g/day to approximately 1 mg/day; approximately 0.001 to approximately 500 ⁇ g/day; approximately 0.001 to approximately 100 ⁇ g/day; approximately 0.025 to approximately 75 ⁇ g/day; approximately 0.025 to approximately 65 ⁇ g/day; approximately 0.025 to approximately 60 ⁇ g/day; approximately 0.025 to approximately 55 ⁇ g/day; approximately 0.025 to approximately 50 ⁇ g/day; approximately 0.025 to approximately 45 ⁇ g/day; approximately 0.025 to approximately 40 ⁇ g/day; approximately 0.025 to approximately 35 ⁇ g/day; approximately 0.005 to approximately 30 ⁇ g/day; approximately 0.005 to approximately 25 ⁇ g/day; approximately 0.005 to approximately 20 ⁇ g/day; and
  • the dosage of sulindac is from approximately 0.01 to approximately 15 ⁇ g/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 10 ⁇ g/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 5 ⁇ g/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 20 ⁇ g/day. In another embodiment, the sulindac is administered in a drug depot that releases 9.6 ⁇ g/day.
  • the anti-inflammatory agent in the depot is clonidine, also referred to as 2,6-dichloro-N-2-imidazolidinyldenebenzenamine.
  • Clonidine or a pharmaceutically acceptable salt thereof is available from various pharmaceutical manufactures.
  • the dosage may be from approximately 0.0005 to approximately 960 ⁇ g/day. Additional dosages of clonidine include from approximately 0.0005 to approximately 900 ⁇ g/day; approximately 0.0005 to approximately 500 ⁇ g/day; approximately 0.0005 to approximately 250 ⁇ g/day; approximately 0.0005 to approximately 100 ⁇ g/day; approximately 0.0005 to approximately 75 ⁇ g/day; approximately 0.001 to approximately 70 ⁇ g/day; approximately 0.001 to approximately 65 ⁇ g/day; approximately 0.001 to approximately 60 ⁇ g/day; approximately 0.001 to approximately 55 ⁇ g/day; approximately 0.001 to approximately 50 ⁇ g/day; approximately 0.001 to approximately 45 ⁇ g/day; approximately 0.001 to approximately 40 ⁇ g/day; approximately 0.001 to approximately 35 ⁇ g/day; approximately 0.0025 to approximately 30 ⁇ g/day; approximately 0.0025 to approximately 25 ⁇ g/day; approximately 0.0025 to approximately 20 ⁇ g/day; approximately 0.0025 to approximately 15 ⁇ g/day; approximately 0.00
  • the dosage of clonidine is from approximately 0.005 to approximately 15 ⁇ g/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to approximately 10 ⁇ g/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to approximately 5 ⁇ g/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to 2.5 ⁇ g/day. In some embodiments, the amount of clonidine is between 40 and 600 ⁇ g/day. In some embodiments, the amount of clonidine is between 200 and 400 ⁇ g/day.
  • the anti-inflammatory agent in the drug depot comprises fluocinolone or a pharmaceutically acceptable salt thereof such as the acetonide salt.
  • Fluocinolone is available from various pharmaceutical manufacturers.
  • the dosage of fluocinolone may be from approximately 0.0005 to approximately 100 ⁇ g/day.
  • Additional dosages of fluocinolone include from approximately 0.0005 to approximately 50 ⁇ g/day; approximately 0.0005 to approximately 25 ⁇ g/day; approximately 0.0005 to approximately 10 ⁇ g/day; approximately 0.0005 to approximately 5 ⁇ g/day; approximately 0.0005 to approximately 1 ⁇ g/day; approximately 0.0005 to approximately 0.75 ⁇ g/day; approximately 0.0005 to approximately 0.5 ⁇ g/day; approximately 0.0005 to approximately 0.25 ⁇ g/day; approximately 0.0005 to approximately 0.1 ⁇ g/day; approximately 0.0005 to approximately 0.075 ⁇ g/day; approximately 0.0005 to approximately 0.05 ⁇ g/day; approximately 0.001 to approximately 0.025 ⁇ g/day; approximately 0.001 to approximately 0.01 ⁇ g/day; approximately 0.001 to approximately 0.0075 ⁇ g/day; approximately 0.001 to approximately 0.005 ⁇ g/day; approximately 0.001 to approximately 0.025 ⁇ g/day; and approximately 0.002 ⁇ g/day.
  • the dosage of fluocinolone is from approximately 0.001 to approximately 15 ⁇ g/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to approximately 10 ⁇ g/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to approximately 5 ⁇ g/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to 2.5 ⁇ g/day. In some embodiments, the amount of fluocinolone is between 40 and 600 ⁇ g/day. In some embodiments, the amount of fluocinolone is between 200 and 400 ⁇ g/day.
  • the anti-inflammatory agent in the drug depot is dexamethasone free base or dexamethasone acetate, also referred to as 8S,9R,10S,11S,13S,14S,16R,17R)-9-Fluoro-11,17-dihydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,11,12,14,15,16 octahydrocyclopenta[a]-phenanthren-3-one), or a pharmaceutically acceptable salt thereof, which is available from various manufacturers.
  • dexamethasone may be released from the depot at a dose of about 10 pg to about 80 mg/day, about 2.4 ng/day to about 50 mg/day, about 50 ng/day to about 2.5 mg/day, about 250 ng/day to about 250 ug/day, about 250 ng/day to about 50 ug/day, about 250 ng/day to about 25 ug/day, about 250 ng/day to about 1 ug/day, about 300 ng/day to about 750 ng/day or about 0.50 ug/day.
  • the dose may be about 0.01 to about 10 ⁇ g/day or about 1 ng to about 120 ⁇ g/day.
  • the dexamethasone is dexamethasone sodium phosphate.
  • the therapeutic agent in the drug depot is GED (guanidinoethyldisulfide), which is an inducible nitric oxide synthase inhibitor having anti-inflammatory properties.
  • GED may be in its hydrogen carbonate salt form.
  • the dosage of GED may be from approximately 0.0005 ⁇ g/day to approximately 100 mg/day. Additional dosages of GED include from approximately 0.0005 ⁇ g/day to approximately 50 mg/day; approximately 0.0005 ⁇ g/day to approximately 10 mg/day; approximately 0.0005 ⁇ g/day to approximately 1 mg/day; approximately 0.0005 to approximately 800 ⁇ g/day; approximately 0.0005 to approximately 50 ⁇ g/day; approximately 0.001 to approximately 45 ⁇ g/day; approximately 0.001 to approximately 40 ⁇ g/day; approximately 0.001 to approximately 35 ⁇ g/day; approximately 0.0025 to approximately 30 ⁇ g/day; approximately 0.0025 to approximately 25 ⁇ g/day; approximately 0.0025 to approximately 20 ⁇ g/day; and approximately 0.0025 to approximately 15 ⁇ g/day.
  • the dosage of GED is from approximately 0.005 to approximately 15 ⁇ g/day. In another embodiment, the dosage of GED is from approximately 0.005 to approximately 10 ⁇ g/day. In another embodiment, the dosage of GED is from approximately 0.005 to approximately 5 ⁇ g/day. In another embodiment, the dosage of GED is from approximately 0.005 to 2.5 ⁇ g/day. In some embodiments, the amount of GED is between 40 and 600 ⁇ g/day. In some embodiments, the amount of GED is between 200 and 400 ⁇ g/day.
  • the dosage of GED is between 0.5 and 4 mg/day. In another exemplary embodiment the dosage of GED is between 0.75 and 3.5 mg/day.
  • the anti-inflammatory agent in the depot comprises lovastatin.
  • Lovastatin is a statin that may be obtained from various manufacturers in various forms (e.g., injection, powder, etc.).
  • lovastatin may be obtained from Merck as Mevacor® (see U.S. Pat. No. 4,231,938, the entire disclosure is herein incorporated by reference).
  • Suitable pharmaceutically acceptable salts of lovastatin include one or more compounds derived from bases such as sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, 1-deoxy-2-(methylamino)-D-glucitol, magnesium hydroxide, zinc hydroxide, aluminum hydroxide, ferrous or ferric hydroxide, ammonium hydroxide or organic amines such as N-methylglucamine, choline, arginine or the like or combinations thereof.
  • Suitable pharmaceutically acceptable salts of lovastatin include lithium, calcium, hemicalcium, sodium, potassium, magnesium, aluminum, ferrous or ferric salts thereof or a combination thereof.
  • the therapeutically effective amount of lovastatin comprises from about 0.1 pg to about 2000 mg, for example, 0.1 ng to 1000 mg, 500 mg, 100 mg, 50 mg, 25 mg, 10 mg, 1 mg, 50 ⁇ g, 25 ⁇ g, 10 ⁇ g, 1 ⁇ g, 500 ng, 250 ng, 100 ng, 75 ng, 50 ng, 25 ng, 15 ng, 10 ng, 5 ng, or 1 ng of lovastatin per day.
  • the dosage may be, for example from about 3 ng/day to 0.3 ⁇ g/day.
  • the analgesic agent in the drug depot is morphine.
  • Morphine is also referred to as (5 ⁇ ,6 ⁇ )-7,8-didehydro-4,5-epoxy-17-methylmorphinan-3,6-diol and has the chemical formula C 17 H 19 NO 3 .
  • Morphine or a pharmaceutically acceptable salt thereof is available from various manufacturers.
  • the morphine comprises morphine sulfate or hydrochloride.
  • the dosage of the morphine may be from 0.1 mg to 1000 mg per day.
  • the dosage of morphine may be for example, 0.1 mg to 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg of morphine per day.
  • the analgesic agent in the drug depot is tramadol.
  • Tramadol is also referred to as ( ⁇ )cis-2-[(dimethylamino)methyl]-1-(3-methoxyphenyl)cyclohexanol hydrochloride and has the chemical formula C 16 H 25 NO 2 .
  • Tramadol or a pharmaceutically acceptable salt thereof is available from various manufacturers.
  • tramadol HCL was used.
  • the dosage of the tramadol may be from 0.01 mg to 500 mg per day.
  • the dosage of tramadol may be for example, 0.1 mg to 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, or 500 mg of tramadol per day.
  • the drug depot contains sufficient tramadol to release between 2.5 and 30 mg/kg/day. In another embodiment the drug depot contains sufficient tramadol to release between 3 and 27.5 mg/kg/day.
  • the at least one anti-inflammatory agent and at least one analgesic agent may also be administered with non-active ingredients. These non-active ingredients may have multi-functional purposes including the carrying, stabilizing and controlling the release of the therapeutic agent(s).
  • the sustained release process may be by a solution-diffusion mechanism or it may be governed by an erosion-sustained process.
  • the depot will be a solid or semi-solid formulation comprised of a biocompatible material that can be biodegradable.
  • solid is intended to mean a rigid material, while “semi-solid” is intended to mean a material that has some degree of flexibility, thereby allowing the depot to bend and conform to the surrounding tissue requirements.
  • the non-active ingredients will be durable within the tissue site for a period of time equal to (for biodegradable components) or greater than (for non-biodegradable components) the planned period of drug delivery.
  • the depot material may have a melting point or glass transition temperature close to or higher than body temperature, but lower than the decomposition or degradation temperature of the therapeutic agent.
  • the pre-determined erosion of the depot material can also be used to provide for slow release of the loaded therapeutic agent(s).
  • the drug depot may not be biodegradable.
  • the drug depot may comprise polyurethane, polyurea, polyether(amide), PEBA, thermoplastic elastomeric olefin, copolyester, and styrenic thermoplastic elastomer, steel, aluminum, stainless steel, titanium, metal alloys with high non-ferrous metal content and a low relative proportion of iron, carbon fiber, glass fiber, plastics, ceramics or combinations thereof.
  • these types of drug depots may need to be removed.
  • the depot may comprise a biodegradable material.
  • a biodegradable material There are numerous materials available for this purpose and having the characteristic of being able to breakdown or disintegrate over a prolonged period of time when positioned at or near the target tissue.
  • the mechanism of the degradation process can be hydrolytical or enzymatical in nature, or both.
  • the degradation can occur either at the surface (heterogeneous or surface erosion) or uniformly throughout the drug delivery system depot (homogeneous or bulk erosion).
  • the depot may comprise a bioabsorbable, and/or a biodegradable biopolymer that may provide immediate release, or sustained release of the at least one analgesic agent and at least one anti-inflammatory agent.
  • suitable sustained release biopolymers include but are not limited to poly (alpha-hydroxy acids), poly (lactide-co-glycolide) (PLGA or PLG), polylactide (PLA), polyglycolide (PG), polyethylene glycol (PEG) conjugates of poly (alpha-hydroxy acids), polyorthoesters, polyaspirins, polyphosphagenes, collagen, starch, pre-gelatinized starch, hyaluronic acid, chitosans, gelatin, alginates, albumin, fibrin, vitamin E analogs, such as alpha tocopheryl acetate, d-alpha tocopheryl succinate, D,L-lactide, or L-lactide, -caprolactone, dextrans, vinylpyr
  • the depot comprises 50:50 PLGA to 100 PLA.
  • the molecular weight range is 0.45 to 0.8 dI/g.
  • the molecular weight of the polymer can be a wide range of values.
  • the average molecular weight of the polymer can be from about 1000 to about 10,000,000; or about 1,000 to about 1,000,000; or about 5,000 to about 500,000; or about 10,000 to about 100,000; or about 20,000 to 50,000.
  • the at least one biodegradable polymer comprises poly(lactic-co-glycolic acid) (PLA) or poly(orthoester) (POE) or a combination thereof.
  • the poly(lactic-co-glycolic acid) may comprise a mixture of polyglycolide (PGA) and polylactide and in some embodiments, in the mixture, there is more polylactide than polyglycolide.
  • polylactide and 0% polyglycolide there is 100% polylactide and 0% polyglycolide; 95% polylactide and 5% polyglycolide; 90% polylactide and 10% polyglycolide; 85% polylactide and 15% polyglycolide; 80% polylactide and 20% polyglycolide; 75% polylactide and 25% polyglycolide; 70% polylactide and 30% polyglycolide; 65% polylactide and 35% polyglycolide; 60% polylactide and 40% polyglycolide; 55% polylactide and 45% polyglycolide; 50% polylactide and 50% polyglycolide; 45% polylactide and 55% polyglycolide; 40% polylactide and 60% polyglycolide; 35% polylactide and 65% polyglycolide; 30% polylactide and 70% polyglycolide; 25% polylactide and 75% polyglycolide; 20% polylactide and 80% polyglycolide; 15% polyl
  • polylactide there is at least 95% polylactide; at least 90% polylactide; at least 85% polylactide; at least 80% polylactide; at least 75% polylactide; at least 70% polylactide; at least 65% polylactide; at least 60% polylactide; at least 55%; at least 50% polylactide; at least 45% polylactide; at least 40% polylactide; at least 35% polylactide; at least 30% polylactide; at least 25% polylactide; at least 20% polylactide; at least 15% polylactide; at least 10% polylactide; or at least 5% polylactide; and the remainder of the biopolymer being polyglycolide.
  • the drug depot comprises poly(lactide-co-glycolide) (PLGA), polylactide (PLA), polyglycolide (PGA), D-lactide, D,L-lactide, L-lactide, D,L-lactide- ⁇ -caprolactone, D,L-lactide-glycolide- ⁇ -caprolactone, glycolide-caprolactone or a combination thereof.
  • PLGA poly(lactide-co-glycolide)
  • PLA polylactide
  • PGA polyglycolide
  • D-lactide D,L-lactide, L-lactide, D,L-lactide- ⁇ -caprolactone, D,L-lactide-glycolide- ⁇ -caprolactone, glycolide-caprolactone or a combination thereof.
  • implantable elastomeric depot compositions having a blend of polymers with different end groups are used the resulting formulation will have a lower burst index and a regulated duration of delivery.
  • polymers with acid e.g., carboxylic acid
  • ester end groups e.g., lauryl, methyl or ethyl ester end groups.
  • the comonomer ratio of the various monomers that form a polymer e.g., the L/G/CL (CL refers to caprolactone, G refers to glycolic acid and L refers to lactic acid) or G/CL ratio for a given polymer
  • L/G/CL caprolactone
  • G glycolic acid
  • L lactic acid
  • G/CL ratio for a given polymer there will be a resulting depot composition having a regulated burst index and duration of delivery.
  • a depot composition having a polymer with a L/G ratio of 50:50 may have a short duration of delivery ranging from about two days to about one month; a depot composition having a polymer with a L/G ratio of 65:35 may have a duration of delivery of about two months; a depot composition having a polymer with a L/G ratio of 75:25 or L/CL ratio of 75:25 may have a duration of delivery of about three months to about four months; a depot composition having a polymer ratio with a L/G ratio of 85:15 may have a duration of delivery of about five months; a depot composition having a polymer with a L/CL ratio of 25:75 or PLA may have a duration of delivery greater than or equal to six months; a depot composition having a terpolymer of CL/G/L with G greater than 50% and L greater than 10% may have a duration of delivery of about one month and a depot composition having a terpolymer of CL/G/L with G less than 50% and L less less than
  • the biodegradable polymer comprises at least 10 wt %, at least 50 wt. %, at least 60 wt. %, at least 70 wt. %, at least 80 wt. %, at least 85 wt. %, at least 90 wt. %, at least 95 wt. %, or at least 99 wt. % of the formulation.
  • the at least one biodegradable polymer and the analgesic and the anti-inflammatory are the only components of the pharmaceutical formulation.
  • At least 75% of the particles have a size from about 1 micrometer to about 200 micrometers. In some embodiments, at least 85% of the particles have a size from about 1 micrometer to about 100 micrometers. In some embodiments, at least 95% of the particles have a size from about 5 micrometer to about 30 micrometers. In some embodiments, all of the particles have a size from about 10 micrometer to about 30 micrometers.
  • At least 75% of the particles have a size from about 5 micrometer to about 20 micrometers. In some embodiments, at least 85% of the particles have a size from about 5 micrometers to about 20 micrometers. In some embodiments, at least 95% of the particles have a size from about 5 micrometer to about 20 micrometers. In some embodiments, all of the particles have a size from about 5 micrometer to about 20 micrometers.
  • the depot may optionally contain inactive materials such as buffering agents and pH adjusting agents such as potassium bicarbonate, potassium carbonate, potassium hydroxide, sodium acetate, sodium borate, sodium bicarbonate, sodium carbonate, sodium hydroxide or sodium phosphate; degradation/release modifiers; drug release adjusting agents; emulsifiers; preservatives such as benzalkonium chloride, chlorobutanol, phenylmercuric acetate and phenylmercuric nitrate, sodium bisulfite, sodium bisulfate, sodium thiosulfate, thimerosal, methylparaben, polyvinyl alcohol and phenylethyl alcohol; solubility adjusting agents; stabilizers; and/or cohesion modifiers.
  • buffering agents and pH adjusting agents such as potassium bicarbonate, potassium carbonate, potassium hydroxide, sodium acetate, sodium borate, sodium bicarbonate, sodium carbonate, sodium hydroxide or sodium phosphate
  • degradation/release modifiers
  • any such inactive materials will be present within the range of 0-75 wt %, and more typically within the range of 0-30 wt %.
  • the depot may comprise sterile preservative free material.
  • the depot can be different sizes, shapes and configurations. There are several factors that can be taken into consideration in determining the size, shape and configuration of the drug depot. For example, both the size and shape may allow for ease in positioning the drug depot at the target tissue site that is selected as the implantation or injection site. In addition, the shape and size of the system should be selected so as to minimize or prevent the drug depot from moving after implantation or injection. In various embodiments, the drug depot can be shaped like a flat surface such as a disc, film, strip or sheet or the like. Flexibility may be a consideration so as to facilitate placement of the drug depot.
  • the drug depot can be different sizes, for example, the drug depot may be a length of from about 0.5 mm to 5 mm and have a diameter of from about 0.01 to about 2 mm. In various embodiments, the drug depot may have a layer thickness of from about 0.005 to 1.0 mm, such as, for example, from 0.05 to 0.75 mm.
  • the drug depot may be in the form of a film, patch or strip and may have a thickness of about 500 microns to about 5,000 microns or in some embodiments about 0.1 mm to about 3 mm or in some embodiments about 0.1 mils to about 60 mils.
  • the drug depot when the drug depot comprises a film or strip, it may be placed at the incision site before the site is closed.
  • the film or strip may for example be made of thermoplastic materials. Additionally, specific materials that may be advantageous for use in the film or strip include but are not limited to the compounds identified above as sustained release biopolymers.
  • the drug depot may be formed by mixing the at least one analgesic agent and the at least one anti-inflammatory agent with the polymer.
  • Radiographic markers can be included on the drug depot to permit the user to position the depot accurately into the target site of the patient. These radiographic markers will also permit the user to track movement and degradation of the depot at the site over time. In this embodiment, the user may accurately position the depot in the site using any of the numerous diagnostic imaging procedures. Such diagnostic imaging procedures include, for example, X-ray imaging or fluoroscopy. Examples of such radiographic markers include, but are not limited to, barium, calcium phosphate, and/or metal beads or particles. In various embodiments, the radiographic marker could be a spherical shape or a ring around the depot.
  • the drug depot can be part of a packing material or several drug depots can placed on or around each other until the target area is packed.
  • the packing material can act as a hemostatic agent and absorb bodily fluid and/or blood during or after surgery.
  • Packing material includes cellulose based materials, such as cellulose gauze made from cotton or regenerated cellulose fiber, regenerated cellulose sponge, other cellulose fibers or the like can be utilized to absorb body fluids and blood during surgery.
  • the packing material can also contain thrombin, chitin, chitosan, fibrin, amorphous fumed silica, gelatin, corn starch, collagen, polyethylene film, polyvinyl acetate, ethylene-vinyl acetate copolymers, metal foils, laminates of cloth or paper, or a plastic film such as for example, resin-like polyethylene, vinyl acetate homopolymers, and ethylene-vinyl acetate, polypropylene, polyesters, PVC, polysaccharides, hyaluronic acid, xanthan, galactomannan, alginate, non-woven fabrics, or the like.
  • the packing material reduces risk of contamination or infection and reduces the risk of interfering with the wound healing process and hinder the prompt recovery of the patient.
  • packing involves the application of packing material, such as for example, gauze, gel, cotton balls, cotton wedges, sponge, or the like to the target tissue site (e.g., nasal and/or sinus cavity, or cardiac tissue).
  • the drug depot may be placed at or near the target tissue site alone or several drug depots can be packed at the target tissue site.
  • the drug depot can be placed at the target tissue site and be packed in by the packing material (e.g., gauze, gel, cotton balls, cotton wedges, etc.) so that the drug depot remains at or near the target tissue site.
  • the packing material reduces or prevents the drug depot from migrating away from the implant site prior to surgical closure.
  • the drug depot will release the therapeutic agent locally at the site of implantation.
  • the packing material may be coated with a therapeutic agent (e.g., antibiotics, petrolatum, etc.).
  • a therapeutic agent e.g., antibiotics, petrolatum, etc.
  • the packing material can be placed into the nose one layer at a time, folding one layer on top of the other until the area is completely packed.
  • the end of the nose may be taped to keep the packing material in place or to prevent the patient from pulling it out.
  • the packing material may be left at the site or degrade over time or it may be removed within 24-48 hours following surgery.
  • the packing material can abut firm tissue so that tape is not needed.
  • turbinates are folds of tissue on the insides of the nose. The folds are sufficiently firm to support packing.
  • a piece of gauze or cotton is wedged with the drug depot (e.g., film, strip, etc.) in between the turbinates where the drug depot will release the therapeutic agent in the adjacent blood vessels.
  • FIG. 1A illustrates a magnified top view of one embodiment of the implantable drug depot in the form of a film or strip 10 that has the analgesic and/or an anti-inflammatory agent 12 disposed on or in the film or strip.
  • the therapeutic agent e.g., analgesic and/or anti-inflammatory
  • FIG. 1B illustrates a magnified top view of one embodiment of the implantable drug depot in the form of a multiple films or strips 10 that are stacked shown as 17 one on each other that have the analgesic and/or an anti-inflammatory agent 12 disposed on or in the film or strip.
  • the films or strips are shown stacked together and can be used as packing material at or near a cardiac tissue or within the nasal or sinus cavity.
  • FIG. 2 illustrates a magnified side view of one embodiment of the implantable drug depot 10 in the form of a sponge 19 that has the analgesic and/or an anti-inflammatory agent 12 disposed on or in the sponge.
  • the therapeutic agent e.g., analgesic and/or anti-inflammatory
  • FIG. 3 illustrates a partial, side sectional view of a human head illustrating the parts of the nasal cavity 14 with the drug depot in the form of a film or strip 10 on packing material 13 administered thereto.
  • the drug depot is held in place and is packed against the anterior nasal cavity against the nasal mucosa, where the therapeutic agent can be released.
  • the packing material holds the drug depot in position at the target tissue site.
  • FIG. 4 illustrates a partial, front sectional view of a human head illustrating portions of a nasal cavity and sinus cavity with the drug depot in the form of a film or strip 11 administered to the sinus frontilis 16 where the therapeutic agent can be released as the film or strip degrades.
  • the therapeutic agent can be released as the film or strip degrades.
  • localized delivery of the therapeutic agent can be accomplished locally in the sinus cavity to prevent, treat or reduce inflammation and/or pain locally at the site of implantation.
  • the sinus frontilis is shown other areas of the sinus cavity can have the drug depot implanted (e.g., sinus sphenoidalis, cellulae ethmoidalis, the sinus maximilaris 18 , etc.).
  • FIG. 5 schematically depicts the heart 20 and the target vessel, which is represented by the descending coronary artery 23 that had a surgical procedure indicated by the suture 24 after myocardial infraction.
  • a plurality of drug depot films or strips 30 and 32 containing an anti-inflammatory or analgesic agent near the surgical site 24 , edema, inflammation and/or pain can be reduced to speed the patient's recovery.
  • the films or strips can be part of packing material that can absorb blood and fluid and degrade over time.
  • the drug depot in the form of a film or strip can be placed in or around cardiac tissue.
  • the drug depot in the form of a film or strip can be placed at or near the left coronary artery 21 , circumflex artery 22 , aorta 25 , and right coronary artery 26 or other areas at or near the heart.
  • the drug depot may be implanted at, near or in cardiac tissue, such as for example, pericardium (e.g., serous pericardium, parietal pericardium, fibrous pericardium, visceral pericardium) myocardium, epicardium, or muscle, connective tissue at or near the heart or like tissue.
  • pericardium e.g., serous pericardium, parietal pericardium, fibrous pericardium, visceral pericardium
  • the drug depot comprises a gel that can be placed at the target tissue site as a film.
  • the gel has a pre-dosed viscosity in the range of about 1 to about 500 centipoise (cps), 1 to about 200 cps, or 1 to about 100 cps.
  • cps centipoise
  • the viscosity of the gel will increase and the gel will have a modulus of elasticity (Young's modulus) in the range of about 1 ⁇ 10 4 to about 6 ⁇ 10 5 dynes/cm 2 , or 2 ⁇ 10 4 to about 5 ⁇ 10 5 dynes/cm 2 , or 5 ⁇ 10 4 to about 5 ⁇ 10 5 dynes/cm 2 .
  • a depot comprises an adherent gel comprising at least one analgesic agent and at least one anti-inflammatory agent that is evenly distributed throughout the gel.
  • the gel may be of any suitable type, as previously indicated, and should be sufficiently viscous so as to prevent the gel from migrating from the targeted delivery site once deployed; the gel should, in effect, “stick” or adhere to the targeted tissue site.
  • the gel may, for example, solidify upon contact with the targeted tissue or after deployment from a targeted delivery system.
  • the targeted delivery system may be, for example, a syringe, a catheter, needle or cannula or any other suitable device.
  • the targeted delivery system may inject the gel into or on the targeted tissue site.
  • the therapeutic agent may be mixed into the gel prior to the gel being deployed at the targeted tissue site.
  • the gel may be part of a two-component delivery system and when the two components are mixed, a chemical process is activated to form the gel and cause it to stick or to adhere to the target tissue.
  • a gel that hardens or stiffens after delivery.
  • hardening gel formulations may have a pre-dosed modulus of elasticity in the range of about 1 ⁇ 10 4 to about 3 ⁇ 10 5 dynes/cm 2 , or 2 ⁇ 10 4 to about 2 ⁇ 10 5 dynes/cm 2 , or 5 ⁇ 10 4 to about 1 ⁇ 10 5 dynes/cm 2 .
  • the post-dosed hardening gels may have a rubbery consistency and have a modulus of elasticity in the range of about 1 ⁇ 10 4 to about 2 ⁇ 10 6 dynes/cm 2 , or 1 ⁇ 10 5 to about 7 ⁇ 10 5 dynes/cm 2 , or 2 ⁇ 10 5 to about 5 ⁇ 10 5 dynes/cm 2 .
  • the polymer concentration may affect the rate at which the gel hardens (e.g., a gel with a higher concentration of polymer may coagulate more quickly than gels having a lower concentration of polymer).
  • the resulting matrix is solid but is also able to conform to the irregular surface of the tissue (e.g., recesses and/or projections in tissue).
  • the percentage of polymer present in the gel may also affect the viscosity of the polymeric composition. For example, a composition having a higher percentage by weight of polymer is typically thicker and more viscous than a composition having a lower percentage by weight of polymer. A more viscous composition tends to flow more slowly. Therefore, a composition having a lower viscosity may be preferred in some instances.
  • the molecular weight of the gel can be varied by any one of the many methods known in the art.
  • the choice of method to vary molecular weight is typically determined by the composition of the gel (e.g., polymer versus non-polymer).
  • the degree of polymerization can be controlled by varying the amount of polymer initiators (e.g. benzoyl peroxide), organic solvents or activator (e.g. DMPT), crosslinking agents, polymerization agent, and/or reaction time.
  • Suitable gel polymers may be soluble in an organic solvent.
  • the solubility of a polymer in a solvent varies depending on the crystallinity, hydrophobicity, hydrogen-bonding and molecular weight of the polymer. Lower molecular weight polymers will normally dissolve more readily in an organic solvent than high-molecular weight polymers.
  • a polymeric gel which includes a high molecular weight polymer, tends to coagulate or solidify more quickly than a polymeric composition, which includes a low-molecular weight polymer.
  • Polymeric gel formulations, which include high molecular weight polymers also tend to have a higher solution viscosity than a polymeric gel, which include a low-molecular weight polymer.
  • the gel When the gel is designed to be a flowable gel, it can vary from low viscosity, similar to that of water, to a high viscosity, similar to that of a paste, depending on the molecular weight and concentration of the polymer used in the gel.
  • the viscosity of the gel can be varied such that the polymeric composition can be applied to a patient's tissues by any convenient technique, for example, by brushing, dripping, injecting, or painting. Different viscosities of the gel will depend on the technique used to apply the composition.
  • the gel has an inherent viscosity (abbreviated as “I.V.” and units are in deciliters/gram), which is a measure of the gel's molecular weight and degradation time (e.g., a gel with a high inherent viscosity has a higher molecular weight and longer degradation time).
  • I.V inherent viscosity
  • a gel with a high molecular weight provides a stronger film or strip and the film or strip takes more time to degrade.
  • a gel with a low molecular weight degrades more quickly and provides a softer film or strip.
  • the gel has a molecular weight, as shown by the inherent viscosity, from about 0.10 dL/g to about 1.2 dL/g or from about 0.10 dL/g to about 0.40 dL/g.
  • the gel can have a viscosity of about 300 to about 5,000 centipoise (cp). In other embodiments, the gel can have a viscosity of from about 5 to about 300 cps, from about 10 cps to about 50 cps, from about 15 cps to about 75 cps at room temperature.
  • the gel may optionally have a viscosity enhancing agent such as, for example, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl methylcellulose, carboxymethylcellulose and salts thereof, Carbopol, poly-(hydroxyethylmethacrylate), poly-(methoxyethylmethacrylate), poly(methoxyethoxyethyl methacrylate), polymethylmethacrylate (PMMA), methylmethacrylate (MMA), gelatin, polyvinyl alcohols, propylene glycol, PEG 200, PEG 300, PEG 400, PEG 500, PEG 600, PEG 700, PEG 800, PEG 900, PEG 1000, PEG 1450, PEG 3350, PEG 4500, PEG 8000 or combinations thereof.
  • a viscosity enhancing agent such as, for example, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl methylcellulose, carboxymethylcellulose and salts thereof
  • the polymeric composition when a polymer is employed in the gel, includes about 10 wt % to about 90 wt % or about 30 wt % to about 60 wt % of the polymer.
  • the gel is a hydrogel made of high molecular weight biocompatible elastomeric polymers of synthetic or natural origin.
  • a desirable property for the hydrogel to have is the ability to respond rapidly to mechanical stresses, particularly shears and loads, in the human body.
  • Hydrogels obtained from natural sources are particularly appealing because they are more likely to be biodegradable and biocompatible for in vivo applications.
  • Suitable hydrogels include natural hydrogels, such as, for example, gelatin, collagen, silk, elastin, fibrin and polysaccharide-derived polymers like agarose, and chitosan, glucomannan gel, hyaluronic acid, polysaccharides, such as cross-linked carboxyl-containing polysaccharides, or a combination thereof.
  • Synthetic hydrogels include, but are not limited to those formed from polyvinyl alcohol, acrylamides such as polyacrylic acid and poly (acrylonitrile-acrylic acid), polyurethanes, polyethylene glycol (e.g., PEG 3350, PEG 4500, PEG 8000), silicone, polyolefins such as polyisobutylene and polyisoprene, copolymers of silicone and polyurethane, neoprene, nitrile, vulcanized rubber, poly(N-vinyl-2-pyrrolidone), acrylates such as poly(2-hydroxy ethyl methacrylate) and copolymers of acrylates with N-vinyl pyrolidone, N-vinyl lactams, polyacrylonitrile or combinations thereof.
  • polyvinyl alcohol acrylamides such as polyacrylic acid and poly (acrylonitrile-acrylic acid), polyurethanes, polyethylene glycol (e.g., PEG 3350, P
  • the hydrogel materials may further be cross-linked to provide further strength as needed.
  • polyurethanes include thermoplastic or thermoset polyurethanes, aliphatic or aromatic polyurethanes, polyetherurethane, polycarbonate-urethane or silicone polyether-urethane, or a combination thereof.
  • microspheres may be dispersed within the gel, the microspheres being loaded with at least one analgesic agent and/or at least one anti-inflammatory agent.
  • the microspheres provide for a sustained release of the at least one analgesic agent and at least one anti-inflammatory agent.
  • the gel which is biodegradable, prevents the microspheres from releasing the analgesic agent and/or anti-inflammatory agent; the microspheres thus do not release the analgesic agent and/or anti-inflammatory agent until they have been released from the gel.
  • a gel may be deployed around a target tissue site (e.g., a cardiac tissue).
  • microspheres Dispersed within the gel is a plurality of microspheres that encapsulate the desired therapeutic agent. Certain of these microspheres degrade once released from the gel, thus releasing the analgesic agent and/or anti-inflammatory agent.
  • the analgesic agents and/or anti-inflammatory agents may be placed into separate microspheres and then the microspheres combined, or the active ingredients can first be combined and then placed into the microspheres together.
  • Microspheres may disperse relatively quickly, depending upon the surrounding tissue type, and hence disperse the at least one analgesic agent and at least one anti-inflammatory agent.
  • the diameter of the microspheres range from about 10 microns in diameter to about 200 microns in diameter. In some embodiments they range from about 20 to 120 microns in diameters.
  • the present invention also contemplates the use of adherent gels to so constrain dispersal of the therapeutic agent.
  • adherent gels may be deployed, for example, in the sinus cavity, or cardiac tissue, or in surrounding tissue.
  • the depot can be administered to the target site using a “cannula” or “needle” that can be a part of a drug delivery device e.g., a syringe, a gun drug delivery device, or any medical device suitable for the application of a drug to a targeted organ or anatomic region.
  • a drug delivery device e.g., a syringe, a gun drug delivery device, or any medical device suitable for the application of a drug to a targeted organ or anatomic region.
  • the cannula or needle of the drug depot device is designed to cause minimal physical and psychological trauma to the patient.
  • Cannulas or needles include tubes that may be made from materials, such as for example, polyurethane, polyurea, polyether(amide), PEBA, thermoplastic elastomeric olefin, copolyester, and styrenic thermoplastic elastomer, steel, aluminum, stainless steel, titanium, metal alloys with high non-ferrous metal content and a low relative proportion of iron, carbon fiber, glass fiber, plastics, ceramics or combinations thereof.
  • the cannula or needle may optionally include one or more tapered regions.
  • the cannula or needle may be beveled.
  • the cannula or needle may also have a tip style vital for accurate treatment of the patient depending on the site for implantation.
  • tip styles include, for example, Trephine, Cournand, Veress, Huber, Seldinger, Chiba, Francine, Bias, Crawford, deflected tips, Hustead, Lancet, or Tuohey.
  • the cannula or needle may also be non-coring and have a sheath covering it to avoid unwanted needle sticks.
  • the dimensions of the hollow cannula or needle will depend on the site for implantation.
  • the thickness of the cannula or needle will also depend on the site of implantation. In various embodiments, the thickness includes, but is not limited to, from about 0.05 to about 1.655.
  • the gauge of the cannula or needle may be the widest or smallest diameter or a diameter in between for insertion into a human or animal body. The widest diameter is typically about 14 gauge, while the smallest diameter is about 25 gauge. In various embodiments the gauge of the needle or cannula is about 18 to about 22 gauge.
  • the cannula or needle includes dose radiographic markers that indicate location at or near the site beneath the skin, so that the user may accurately position the depot at or near the site using any of the numerous diagnostic imaging procedures.
  • diagnostic imaging procedures include, for example, X-ray imaging or fluoroscopy.
  • radiographic markers include, but are not limited to, barium, calcium phosphate, and/or metal beads or particles.
  • the needle or cannula may include a transparent or translucent portion that can be visualizable by ultrasound, fluoroscopy, x-ray, or other imaging techniques.
  • the transparent or translucent portion may include a radiopaque material or ultrasound responsive topography that increases the contrast of the needle or cannula relative to the absence of the material or topography.
  • the drug depot (e.g., strip, sheet, film, etc.) and/or medical device to administer the drug depot may be sterilizable.
  • one or more components of the drug depot, and/or medical device to administer the drug depot are sterilized by radiation in a terminal sterilization step in the final packaging. Terminal sterilization of a product provides greater assurance of sterility than from processes such as an aseptic process, which require individual product components to be sterilized separately and the final package assembled in a sterile environment.
  • gamma radiation is used in the terminal sterilization step, which involves utilizing ionizing energy from gamma rays that penetrates deeply in the device.
  • Gamma rays are highly effective in killing microorganisms, they leave no residues nor have sufficient energy to impart radioactivity to the device.
  • Gamma rays can be employed when the device is in the package and gamma sterilization does not require high pressures or vacuum conditions, thus, package seals and other components are not stressed.
  • gamma radiation eliminates the need for permeable packaging materials.
  • electron beam (e-beam) radiation may be used to sterilize one or more components of the device.
  • E-beam radiation comprises a form of ionizing energy, which is generally characterized by low penetration and high-dose rates.
  • E-beam irradiation is similar to gamma processing in that it alters various chemical and molecular bonds on contact, including the reproductive cells of microorganisms. Beams produced for e-beam sterilization are concentrated, highly-charged streams of electrons generated by the acceleration and conversion of electricity. E-beam sterilization may be used, for example, when the drug depot is included in a gel.
  • kits may also be used to sterilize the depot and/or one or more components of the device, including, but not limited to, gas sterilization, such as, for example, with ethylene oxide or steam sterilization.
  • gas sterilization such as, for example, with ethylene oxide or steam sterilization.
  • a kit may include additional parts along with the drug depot and/or medical device combined together to be used to implant the drug depot (e.g., film, strip, etc.).
  • the kit may include the drug depot device in a first compartment.
  • the second compartment may include a canister holding the drug depot and any other instruments needed for the localized drug delivery.
  • a third compartment may include gloves, drapes, wound dressings, packing material, and other procedural supplies for maintaining sterility of the implanting process, as well as an instruction booklet.
  • a fourth compartment may include additional cannulas and/or needles.
  • a fifth compartment may include the agent for radiographic imaging. Each tool may be separately packaged in a plastic pouch that is radiation sterilized.
  • a cover of the kit may include illustrations of the implanting procedure and a clear plastic cover may be placed over the compartments to maintain sterility.
  • the analgesic and/or anti-inflammatory may be parenterally administered.
  • parenteral refers to modes of administration, which bypass the gastrointestinal tract, and include for example, intravenous, intramuscular, continuous or intermittent infusion, intraperitoneal, intrasternal, subcutaneous, intra-operatively, intrathecally, intradiscally, peridiscally, epidurally, perispinally, intranasally, intraarticular injection or combinations thereof.
  • a method for delivering a therapeutic agent into a surgery site of a patient comprising inserting a cannula at or near a target tissue site and implanting the drug depot at the target site beneath the skin of the patient and brushing, dripping, spraying, injecting, or painting the gel in the target site to hold or have the drug depot adhere to the target site. In this way unwanted migration of the drug depot away from the target site is reduced or eliminated.
  • therapeutically effective doses may be less than doses administered by other routes (oral, topical, etc.).
  • the drug dose delivered from the drug depot may be, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, and 95% less than the oral dosage or injectable dose.
  • systemic side effects such as for example, liver transaminase elevations, hepatitis, liver failure, myopathy, constipation, etc. may be reduced or eliminated.
  • first the cannula or needle can be inserted through the skin and soft tissue down to the target tissue site and the gel administered (e.g., brushed, dripped, injected, or painted, etc.) at or near the target site.
  • the cannula or needle can be inserted through the skin and soft tissue down to the site of injection and one or more base layer(s) of gel can be administered to the target site.
  • the drug depot can be implanted on or in the base layer(s) so that the gel can hold the depot in place or reduce migration.
  • a subsequent layer or layers of gel can be applied on the drug depot to surround the depot and further hold it in place.
  • the drug depot may be implanted first and then the gel placed (e.g., brushed, dripped, injected, or painted, etc.) around the drug depot to hold it in place.
  • the gel may also avoid the need to suture the drug depot to the target site reducing physical and psychological trauma to the patient.
  • the at least one analgesic and/or anti-inflammatory agent formulation may be used to form different pharmaceutical preparations (e.g., drug depot film, sheet, strip, etc.).
  • the pharmaceutical preparations may be formed in an administration with a suitable pharmaceutical carrier that may be solid or liquid, and placed in the appropriate form for parenteral or other administration as desired.
  • suitable pharmaceutical carrier include but are not limited to water, gelatin, lactose, starches, stearic acid, magnesium stearate, sicaryl alcohol, talc, vegetable oils, benzyl alcohols, gums, waxes, propylene glycol, polyalkylene glycols and other known carriers.
  • Another embodiment provides a method for treating a mammal suffering from pain and/or inflammation, said method comprising administering a therapeutically effective amount of the analgesic and/or anti-inflammatory agent at a target site beneath the skin at or near the target site.
  • the therapeutically effective dosage amount (e.g., analgesic and/or anti-inflammatory) and the release rate profile are sufficient to reduce inflammation and/or pain for a period of at least one day, for example, 1-90 days, 1-10 days, 1-3 days, 3-7 days, 3-12 days; 3-14 days, 7-10 days, 7-14 days, 7-21 days, 7-30 days, 7-50 days, 7-90 days, 7-140 days, 14-140 days, 3 days to 150 days, or 3 days to 6 months.
  • the at least one analgesic and/or anti-inflammatory or a portion of the at least one analgesic and/or anti-inflammatory is administered as a bolus dose at the target tissue to provide an immediate release of the analgesic and/or anti-inflammatory.
  • composition useful for the treatment of inflammation comprising an effective amount of at least one analgesic and/or anti-inflammatory that is capable of being locally administered to a target tissue site.
  • analgesic and/or anti-inflammatory that is capable of being locally administered to a target tissue site.
  • they may be administered locally to the nasal, sinus, and/or cardiac tissue.
  • the at least one analgesic and/or anti-inflammatory is administered parenterally, e.g., by injection.
  • the injection is intra-cardiac, which refers to an injection into the cardiac tissue.
  • An injection may also be into a muscle or other tissue.
  • the analgesic and/or anti-inflammatory is administered by placement into an open patient cavity during surgery.
  • the formulation is implantable into a surgical site at the time of surgery.
  • the active ingredients may then be released from the depot via diffusion in a sustained fashion over a period of time, e.g., 3-15 days, 5-10 days or 7-10 days post surgery in order to address pain and inflammation.
  • the active ingredient may provide longer duration of pain and/or inflammation relief for chronic diseases/conditions with release of one or more drugs up to 6 months or 1 year (e.g., 90, 100, 150, 180 days or longer).
  • the drug depot may release 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof relative to a total amount of at least one analgesic and/or anti-inflammatory agent loaded in the drug depot over a period of 3 to 12 days, 5 to 10 days or 7 to 10 days after the drug depot is administered to the target tissue site.
  • the active ingredient may provide longer duration of pain and/or inflammation relief for chronic diseases/conditions as discussed above with release of one or more drugs up to 6 months or 1 year (e.g., 90, 100, 150, 180 days or longer).
  • an implantable drug depot useful for reducing, preventing or treating pain and/or inflammation is provided in a patient in need of such treatment, the implantable drug depot comprising a therapeutically effective amount of a analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salts thereof, the depot being implantable at a site beneath the skin to reduce, prevent or treat pain and/or inflammation, wherein the drug depot (i) comprises one or more immediate release layer(s) that is capable of releasing about 5% to about 20% of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salts thereof relative to a total amount of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof loaded in the drug depot over a first period of up to 48 hours and (ii) one or more sustain release layer(s) that is capable of releasing about 21% to about 99% of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof relative to a total amount of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt
  • the target tissue site may comprise at least one sinus cavity, nasal cavity, cardiac tissue, muscle, ligament, tendon, cartilage, spinal disc, spinal foraminal space near the spinal nerve root, facet or spinal canal.
  • the inflammation may be associated with orthopedic or spine surgery or a combination thereof.
  • the surgery may be arthroscopic surgery, an excision of a mass, hernia repair, spinal fusion, thoracic, cervical, or lumbar surgery, pelvic surgery or a combination thereof.
  • the active ingredient may provide longer duration of pain and/or inflammation relief for chronic diseases/conditions as discussed above with release of one or more drugs up to 6 months or 1 year (e.g., 90, 100, 150, 180 days or longer).
  • the at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof is encapsulated in a plurality of depots comprising microparticles, microspheres, microcapsules, and/or microfibers suspended in a gel.
  • a method of inhibiting pain and/or inflammation in a patient in need of such treatment, the method comprising delivering one or more biodegradable drug depots comprising a therapeutically effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof to a target tissue site beneath the skin before, during or after surgery, wherein the drug depot releases an effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of 3 days to 6 months.
  • an implantable drug depot useful for preventing or treating pain and/or inflammation in a patient in need of such treatment comprising a therapeutically effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof, the depot being implantable at a site beneath the skin to prevent or treat inflammation, wherein the drug depot releases an effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of 33 days to 6 months.
  • an implantable drug depot comprising one or more immediate release layer(s) that releases a bolus dose of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof at a site beneath the skin and (ii) one or more sustain release layer(s) that releases an effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of 3 to 12 days or 5 to 10 days or 7 to 10 days or 3 days to 6 months.
  • the one or more immediate release layer(s) may comprise poly (lactide-co-glycolide) (PLGA) and the one or more sustain release layer(s) may comprise polylactide (PLA).
  • an implantable drug depot useful for reducing, preventing or treating pain and inflammation in a patient is provided, the implantable drug depot in the form of a film or strip comprising a therapeutically effective amount of an analgesic and an anti-inflammatory agent or pharmaceutically acceptable salts thereof and a polymer; wherein the drug depot is implantable at a site beneath the skin to reduce, prevent or treat pain and inflammation, and the depot is capable of releasing (i) about 5% to about 20% of the analgesic or pharmaceutically acceptable salt thereof relative to a total amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof loaded in the drug depot over a first period of up to 72 hours and (ii) about 21% to about 99% of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof relative to a total amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof loaded in the drug depot over a subsequent period of up to 2 weeks.
  • the drug depot comprising the active ingredients can be made by combining a biocompatible polymer and a therapeutically effective amount of the active ingredients or pharmaceutically acceptable salts thereof and forming the implantable drug depot (e.g., strip, sheet, film) from the combination.
  • a solvent system is typically selected that contains one or more solvent species.
  • the solvent system is generally a good solvent for at least one component of interest, for example, biocompatible polymer and/or therapeutic agent.
  • the particular solvent species that make up the solvent system can also be selected based on other characteristics, including drying rate and surface tension.
  • Solution processing techniques include solvent casting techniques, spin coating techniques, web coating techniques, solvent spraying techniques, dipping techniques, techniques involving coating via mechanical suspension, including air suspension (e.g., fluidized coating), ink jet techniques and electrostatic techniques. Where appropriate, techniques such as those listed above can be repeated or combined to build up the depot to obtain the desired release rate and desired thickness.
  • a solution containing solvent and biocompatible polymer are combined and placed in a mold of the desired size and shape.
  • polymeric regions including barrier layers, therapeutic layers, and so forth can be formed.
  • the solution can further comprise, one or more of the following: other therapeutic agent(s) and other optional additives such as radiographic agent(s), etc. in dissolved or dispersed form. This results in a polymeric matrix region containing these species after solvent removal.
  • a solution containing solvent with dissolved or dispersed therapeutic agent is applied to a pre-existing polymeric region, which can be formed using a variety of techniques including solution processing and thermoplastic processing techniques, whereupon the therapeutic agent is imbibed into the polymeric region.
  • Thermoplastic processing techniques for forming the depot or portions thereof include molding techniques (for example, injection molding, rotational molding, and so forth), extrusion techniques (for example, extrusion, co-extrusion, multi-layer extrusion, and so forth) and casting.
  • Thermoplastic processing in accordance with various embodiments comprises mixing or compounding, in one or more stages, the biocompatible polymer(s) and one or more of the following: the active ingredients, optional additional therapeutic agent(s), radiographic agent(s), and so forth.
  • the resulting mixture is then shaped into an implantable drug depot.
  • the mixing and shaping operations may be performed using any of the conventional devices known in the art for such purposes.
  • thermoplastic processing there exists the potential for the therapeutic agent(s) to degrade, for example, due to elevated temperatures and/or mechanical shear that are associated with such processing.
  • certain therapeutic agents may undergo substantial degradation under ordinary thermoplastic processing conditions.
  • processing is preferably performed under modified conditions, which prevent the substantial degradation of the therapeutic agent(s).
  • some degradation may be unavoidable during thermoplastic processing, degradation is generally limited to 10% or less.
  • processing conditions that may be controlled during processing to avoid substantial degradation of the therapeutic agent(s) are temperature, applied shear rate, applied shear stress, residence time of the mixture containing the therapeutic agent, and the technique by which the polymeric material and the therapeutic agent(s) are mixed.
  • Mixing or compounding biocompatible polymer with therapeutic agent(s) and any additional additives to form a substantially homogenous mixture thereof may be performed with any device known in the art and conventionally used for mixing polymeric materials with additives.
  • a polymer melt may be formed by heating the biocompatible polymer, which can be mixed with various additives (e.g., therapeutic agent(s), inactive ingredients, etc.) to form a mixture.
  • additives e.g., therapeutic agent(s), inactive ingredients, etc.
  • a common way of doing so is to apply mechanical shear to a mixture of the biocompatible polymer(s) and additive(s).
  • Devices in which the biocompatible polymer(s) and additive(s) may be mixed in this fashion include devices such as single screw extruders, twin screw extruders, banbury mixers, high-speed mixers, ross kettles, and so forth.
  • biocompatible polymer(s) and various additives may be premixed prior to a final thermoplastic mixing and shaping process, if desired (e.g., to prevent substantial degradation of the therapeutic agent among other reasons).
  • a biocompatible polymer is precompounded with a radiographic agent (e.g., radio-opacifying agent) under conditions of temperature and mechanical shear that would result in substantial degradation of the therapeutic agent, if it were present.
  • a radiographic agent e.g., radio-opacifying agent
  • This precompounded material is then mixed with therapeutic agent under conditions of lower temperature and mechanical shear, and the resulting mixture is shaped into the active ingredient containing drug depot.
  • the biocompatible polymer can be precompounded with the therapeutic agent under conditions of reduced temperature and mechanical shear. This precompounded material is then mixed with, for example, a radio-opacifying agent, also under conditions of reduced temperature and mechanical shear, and the resulting mixture is shaped into the drug depot.
  • the conditions used to achieve a mixture of the biocompatible polymer and therapeutic agent and other additives will depend on a number of factors including, for example, the specific biocompatible polymer(s) and additive(s) used, as well as the type of mixing device used.
  • a depot is formed comprising PLGA or PLA polymer, a radio-opacifying agent (e.g., bismuth subcarbonate), and a therapeutic agent prone to degradation by heat and/or mechanical shear (e.g., clonidine),
  • the PGLA or PLA can be premixed with the radio-opacifying agent at temperatures of about, for example, 150° C. to 170° C.
  • the therapeutic agent is then combined with the premixed composition and subjected to further thermoplastic processing at conditions of temperature and mechanical shear that are substantially lower than is typical for PGLA or PLA compositions.
  • barrel temperature, volumetric output are typically controlled to limit the shear and therefore to prevent substantial degradation of the therapeutic agent(s).
  • the therapeutic agent and premixed composition can be mixed/compounded using a twin screw extruder at substantially lower temperatures (e.g., 100-105° C.), and using substantially reduced volumetric output (e.g., less than 30% of full capacity, which generally corresponds to a volumetric output of less than 200 cc/min).
  • this processing temperature is well below the melting points of certain active ingredients, such as an anti-inflammatory and analgesic because processing at or above these temperatures will result in substantial therapeutic agent degradation.
  • the processing temperature will be below the melting point of all bioactive compounds within the composition, including the therapeutic agent. After compounding, the resulting depot is shaped into the desired form, also under conditions of reduced temperature and shear.
  • biodegradable polymer(s) and one or more therapeutic agents are premixed using non-thermoplastic techniques.
  • the biocompatible polymer can be dissolved in a solvent system containing one or more solvent species.
  • Any desired agents for example, a radio-opacifying agent, a therapeutic agent, or both radio-opacifying agent and therapeutic agent
  • Solvent is then removed from the resulting solution/dispersion, forming a solid material.
  • the resulting solid material can then be granulated for further thermoplastic processing (for example, extrusion) if desired.
  • the therapeutic agent can be dissolved or dispersed in a solvent system, which is then applied to a pre-existing drug depot (the pre-existing drug depot can be formed using a variety of techniques including solution and thermoplastic processing techniques, and it can comprise a variety of additives including a radio-opacifying agent and/or viscosity enhancing agent), whereupon the therapeutic agent is imbibed on or in the drug depot.
  • the resulting solid material can then be granulated for further processing, if desired.
  • an extrusion processes may be used to form the drug depot comprising a biocompatible polymer(s), therapeutic agent(s) and radio-opacifying agent(s).
  • Co-extrusion may also be employed, which is a shaping process that can be used to produce a drug depot comprising the same or different layers or regions (for example, a structure comprising one or more polymeric matrix layers or regions that have permeability to fluids to allow immediate and/or sustained drug release).
  • Multi-region depots can also be formed by other processing and shaping techniques such as co-injection or sequential injection molding technology.
  • the depot that may emerge from the thermoplastic processing is cooled.
  • cooling processes include air cooling and/or immersion in a cooling bath.
  • a water bath is used to cool the extruded depot.
  • the immersion time should be held to a minimum to avoid unnecessary loss of therapeutic agent into the bath.
  • immediate removal of water or moisture by use of ambient or warm air jets after exiting the bath will also prevent re-crystallization of the drug on the depot surface, thus controlling or minimizing a high drug dose “initial burst” or “bolus dose” upon implantation or insertion if this is release profile is not desired.
  • the drug depot can be prepared by mixing or spraying the drug with the polymer and then molding the depot to the desired shape.
  • active ingredients are used and mixed or sprayed with the PLGA or PEG550 polymer, and the resulting depot may be formed by extrusion and dried.
  • the drug depot when the drug depot comprises a film or strip, it may be formed into a film or strip by methods such as extrusion, coating, spreading, casting or the like. If a multi-layered film or strip is desired, this may be accomplished by co-extruding more than one combination of components, which may be of the same or different composition. A multi-layered film or strip may also be achieved by coating, spreading, or casting a combination onto an already formed film layer.
  • Coating or casting methods are particularly useful for the purpose of forming the films or strips. Specific examples include reverse roll coating, gravure coating, immersion or dip coating, metering rod or meyer bar coating, slot die or extrusion coating, gap or knife over roll coating, air knife coating, curtain coating, or combinations thereof, especially when a multi-layered films or strips are desired.

Abstract

Effective treatments of pain and/or inflammation are provided. Through the administration of a biodegradable drug depot film, patch, strip or sponge being implantable at or near a cardiac tissue or within a nasal or sinus cavity, one can reduce, prevent or treat pain and/or inflammation.

Description

    BACKGROUND
  • Pain and inflammation relief is of prime importance to anyone treating patients undergoing surgery. Proper pain and inflammation relief imparts significant physiological and psychological benefits to the patient. Not only does effective pain and inflammation relief mean a smoother more pleasant postoperative course (e.g., mood, sleep, quality of life, etc.) with earlier discharge from medical/surgical/outpatient facilities, but it may also reduce the onset of chronic pain and inflammation syndromes (e.g., fibromyalgia, myalgia, etc.).
  • Pain serves a biological function. It often signals the presence of damage or disease within the body and is often accompanied by inflammation (redness, swelling, and/or burning). In the case of postoperative pain and inflammation it may be a result of the surgery, or other treatments such as, for example, management of acute pain following burns or non-surgical trauma. The goal for postoperative pain and inflammation management is to reduce or eliminate pain and inflammation discomfort with medication that cause minimum or no side effects.
  • The site of the surgery has a profound effect upon the degree of postoperative pain and inflammation a patient may suffer. In general, operations on the thorax and upper abdomen are more painful and have more inflammation than operations on the lower abdomen, which in turn are more painful and have more inflammation than peripheral operations on the limbs. However, any operation involving a body cavity, large joint surfaces, the spine or deep tissues should be regarded as painful and have some degree of inflammation. In particular, operations on the thorax or upper abdomen may produce widespread changes in pulmonary function, an increase in abdominal muscle tone and an associated decrease in diaphragmatic function. The result will be an inability to cough and clear secretions, which may lead to lung collapse and pneumonia. Prolonged pain and inflammation can reduce physical activity and lead to venous stasis and an increased risk of deep vein thrombosis and consequently pulmonary embolism. In addition, there can be widespread effects on gut and urinary tract motility, which may lead in turn to postoperative ileus, nausea, vomiting and urinary retention. These problems are unpleasant for the patient and may prolong hospital stay. Most patients who experience moderate to severe post-operative pain and inflammation often require pain and inflammation control at least in the first 3 days after trauma or surgery.
  • One area that is ripe for pain and/or inflammation due to trauma or surgery is the nasal and sinus cavities. Physicians are frequently called upon to treat nasal and sinus cavities as a result of tissue desiccation, trauma, infection, or other nasal and sinus diseases. Another area that is also affected by pain and inflammation is cardiac tissue. For example, during a myocardial infarction, commonly known as a heart attack, the blood supply to part of the heart is interrupted. The resulting ischemia (restriction in blood supply) and oxygen shortage, if left untreated for a sufficient period, can cause damage and/or death (infarction) of heart muscle tissue resulting in pain and inflammation.
  • Many treatment options for nasal, sinus and cardiac tissues involve administering analgesic and/or anti-inflammatory medications by oral and parenteral routes (e.g., intramuscular or intravenous, subcutaneous routes). These routes for drug administration often result in off target effects, which can cause increased adverse side effects.
  • Unfortunately, currently available analgesics and/or anti-inflammatory formulations, although effective for short term relief of pain and/or inflammation, require frequent single dose administration every 4 to 12 hours on an as needed basis. These single dose analgesics and/or anti-inflammatory formulations are inconvenient and may interfere with the patient's postoperative inpatient and/or outpatient daytime activities and nighttime sleep and recovery.
  • New analgesics and/or anti-inflammatory compositions and methods are needed to treat or reduce postoperative pain and/or inflammation at or near cardiac tissue or within the nasal or sinus cavity. New analgesics and/or anti-inflammatory compositions and methods that reliably provide long acting analgesic and anti-inflammatory effects over periods of 3 to 10 days are needed.
  • SUMMARY
  • New compositions and methods are provided that effectively prevent, treat or reduce postoperative pain or inflammation in areas at or near cardiac tissue or within the nasal or sinus cavity. In various embodiments, analgesic and/or anti-inflammatory compositions and methods are provided that have long acting analgesic and/or anti-inflammatory effects over periods of 3 to 10 days in a single drug depot or multiple drug depots. New drug depot films and methods are provided, which can easily allow accurate and precise implantation of a drug depot containing analgesic and/or anti-inflammatory compositions. One advantage of the analgesic and/or anti-inflammatory drug depot compositions and methods is that the drug depot can now be easily delivered to the target tissue site (e.g., nasal, sinus and/or cardiac tissue, surgical wound or incision, etc.) and provide pain relief for 3 to 10 days. In this way, accurate and precise implantation of the drug depot can be accomplished. In some embodiments, the drug depot is in the form of a film and can be locally delivered to the nasal, sinus and/or cardiac tissue by packing the drug depot at the target tissue site.
  • In one embodiment, an implantable drug depot is provided useful for reducing, preventing or treating pain and/or inflammation in a patient in need of such treatment, the implantable drug depot being in the form of a biodegradable film and comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent, the depot being implantable at or near a cardiac tissue or within the nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is capable of releasing an effective amount of the analgesic and/or an anti-inflammatory agent over a period of at least one day.
  • In another embodiment, a method of treating or preventing pain and inflammation in a patient in need of such treatment is provided, the method comprising administering one or more biodegradable drug depots comprising a therapeutically effective amount of an analgesic and an anti-inflammatory agent at or near a cardiac tissue or within the nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is in the form of a biodegradable film or strip that releases an effective amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of at least 1 day.
  • In yet another embodiment, a method is provided for reducing pain and inflammation in a patient in need of such treatment, the method comprising delivering one or more biodegradable drug depots in the form of a biodegradable film comprising a therapeutically effective amount of an analgesic and an anti-inflammatory agent or pharmaceutically acceptable salts thereof at or near a cardiac tissue or within the nasal or sinus cavity of the patient, wherein the drug depot releases an effective amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of at least 1 day.
  • The therapeutic agent may for example, be part of a drug depot. The drug depot may: (i) consist of the analgesic and/or an anti-inflammatory agent and the biodegradable polymer(s); or (ii) consist essentially of the analgesic and/or an anti-inflammatory agent; or (iii) comprise the analgesic and/or an anti-inflammatory agent and one or more other active ingredients, surfactants, excipients or other ingredients or combinations thereof. When there are other active ingredients, surfactants, excipients or other ingredients or combinations thereof in the formulation, in some embodiments these other compounds or combinations thereof comprise less than 20 wt. %, less than 19 wt. %, less than 18 wt. %, less than 17 wt. %, less than 16 wt. %, less than 15 wt. %, less than 14 wt. %, less than 13 wt. %, less than 12 wt. %, less than 11 wt. %, less than 10 wt. %, less than 9 wt. %, less than 8 wt. %, less than 7 wt. %, less than 6 wt. %, less than 5 wt. %, less than 4 wt. %, less than 3 wt. %, less than 2 wt. %, less than 1 wt. % or less than 0.5 wt. %.
  • Additional features and advantages of various embodiments will be set forth in part in the description that follows, and in part will be apparent from the description, or may be learned by practice of various embodiments. The objectives and other advantages of various embodiments will be realized and attained by means of the elements and combinations particularly pointed out in the description and appended claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • In part, other aspects, features, benefits and advantages of the embodiments will be apparent with regard to the following description, appended claims and accompanying drawings where:
  • FIG. 1A illustrates a magnified top view of one embodiment of the implantable drug depot in the form of a film or strip that has the analgesic and/or an anti-inflammatory agent disposed on or in the film or strip.
  • FIG. 1B illustrates a magnified top view of one embodiment of the implantable drug depot in the form of multiple films or strips that have the analgesic and/or an anti-inflammatory agent disposed on or in the films or strips. The films or strips are shown stacked together and can be used as packing material to pack at or near a cardiac tissue or within the nasal or sinus cavity.
  • FIG. 2 illustrates a magnified side view of one embodiment of the implantable drug depot in the form of a sponge that has the analgesic and/or an anti-inflammatory agent disposed on or in the sponge.
  • FIG. 3 illustrates a partial, side sectional view of a human head illustrating the parts of the nasal cavity with the drug depot in the form of a film or strip packing administered thereto.
  • FIG. 4 illustrates a partial, front sectional view of a human head illustrating portions of a nasal cavity and sinus cavity with the drug depot in the form of a film or strip administered to the sinus frontilis.
  • FIG. 5 schematically shows tissues and vessels of the heart where the drug depot in the form of multiple films, or strips, or cardiac patch that can be administered thereto.
  • It is to be understood that the figures are not drawn to scale. Further, the relation between objects in a figure may not be to scale, and may in fact have a reverse relationship as to size. The figures are intended to bring understanding and clarity to the structure of each object shown, and thus, some features may be exaggerated in order to illustrate a specific feature of a structure.
  • DETAILED DESCRIPTION
  • For the purposes of this specification and appended claims, unless otherwise indicated, all numbers expressing quantities of ingredients, percentages or proportions of materials, reaction conditions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
  • Notwithstanding the numerical ranges and parameters set forth herein, the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Moreover, all ranges disclosed herein are to be understood to encompass any and all subranges subsumed therein. For example, a range of “1 to 10” includes any and all subranges between (and including) the minimum value of 1 and the maximum value of 10, that is, any and all subranges having a minimum value of equal to or greater than 1 and a maximum value of equal to or less than 10, e.g., 5.5 to 10.
  • Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying drawings. While the invention will be described in conjunction with the illustrated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents that may be included within the invention as defined by the appended claims.
  • The headings below are not meant to limit the disclosure in any way; embodiments under any one heading may be used in conjunction with embodiments under any other heading.
  • It is noted that, as used in this specification and the appended claims, the singular forms “a,” “an,” and “the,” include plural referents unless expressly and unequivocally limited to one referent. Thus, for example, reference to “a drug depot” includes one, two, three or more drug depots.
  • New compositions and methods are provided that effectively prevent, treat or reduce postoperative pain or inflammation in areas at or near cardiac tissue or within the nasal or sinus cavity. In various embodiments, analgesic and/or anti-inflammatory compositions and methods are provided that have long acting analgesic and/or anti-inflammatory effects over periods of 3 to 10 days in a single drug depot or multiple drug depots. New drug depot films and methods are provided, which can easily allow accurate and precise implantation of a drug depot containing analgesic and/or anti-inflammatory compositions. One advantage of the analgesic and/or anti-inflammatory drug depot compositions and methods is that the drug depot can now be easily delivered to the target tissue site (e.g., nasal, sinus and/or cardiac tissue, surgical wound or incision, etc.) and provide pain relief for 3 to 10 days. In this way, accurate and precise implantation of the drug depot can be accomplished. In some embodiments, the drug depot is in the form of a film and can be locally delivered to the nasal, sinus and/or cardiac tissue by packing the drug depot at the target tissue site.
  • In one embodiment, an implantable drug depot is provided useful for reducing, preventing or treating pain and/or inflammation in a patient in need of such treatment, the implantable drug depot being in the form of a biodegradable film and comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent, the depot being implantable at or near a cardiac tissue or within the nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is capable of releasing an effective amount of the analgesic and/or an anti-inflammatory agent over a period of at least one day.
  • DEFINITIONS
  • It is noted that, as used in this specification and the appended claims, the singular forms “a,” “an,” and “the,” include plural referents unless expressly and unequivocally limited to one referent. Thus, for example, reference to “a drug depot” includes one, two, three or more drug depots.
  • Analgesic refers to an agent or compound that can reduce, relieve or eliminate pain. Examples of analgesic agents include but are not limited to acetaminophen, a local anesthetic, such as for example, lidocaine, bupivacaine, ropivacaine, opioid analgesics such as buprenorphine, butorphanol, dextromoramide, dezocine, dextropropoxyphene, diamorphone, fentanyl, alfentanil, sulfentanil, hydrocodone, hydromorphone, ketobemidone, levomethadyl, levorphanol, meperidine, methadone, morphine, nalbuphine, opium, oxycodone, papavereturn, pentazocine, pethidine, phenoperidine, piritramide, dextropropoxyphene, remifentanil, sulfentanil, tilidine, tramadol, codeine, dihydrocodeine, meptazinol, dezocine, eptazocine, flupirtine or a combination thereof.
  • The phrase “anti-inflammatory agent” refers to an agent or compound that has anti-inflammatory effects. These agents may remedy pain by reducing inflammation. Examples of anti-inflammatory agents include, but are not limited to, a statin, sulindac, sulfasalazine, naroxyn, diclofenac, indomethacin, ibuprofen, flurbiprofen, ketoprofen, aclofenac, aloxiprin, aproxen, aspirin, diflunisal, fenoprofen, mefenamic acid, naproxen, phenylbutazone, piroxicam, meloxicam, salicylamide, salicylic acid, desoxysulindac, tenoxicam, ketoralac, flufenisal, salsalate, triethanolamine salicylate, aminopyrine, antipyrine, oxyphenbutazone, apazone, cintazone, flufenamic acid, clonixeril, clonixin, meclofenamic acid, flunixin, colchicine, demecolcine, allopurinol, oxypurinol, benzydamine hydrochloride, dimefadane, indoxole, intrazole, mimbane hydrochloride, paranylene hydrochloride, tetrydamine, benzindopyrine hydrochloride, fluprofen, ibufenac, naproxol, fenbufen, cinchophen, diflumidone sodium, fenamole, flutiazin, metazamide, letimide hydrochloride, nexeridine hydrochloride, octazamide, molinazole, neocinchophen, nimazole, proxazole citrate, tesicam, tesimide, tolmetin, triflumidate, fenamates (mefenamic acid, meclofenamic acid), nabumetone, celecoxib, etodolac, nimesulide, apazone, gold, tepoxalin; dithiocarbamate, or a combination thereof. Anti-inflammatory agents also include other compounds such as steroids, such as for example, fluocinolone, cortisol, cortisone, hydrocortisone, fludrocortisone, prednisone, prednisolone, methylprednisolone, triamcinolone, betamethasone, dexamethasone, beclomethasone, fluticasone interleukin-1 receptor antagonists, thalidomide (a TNF-α release inhibitor), thalidomide analogues (which reduce TNF-α production by macrophages), bone morphogenetic protein (BMP) type 2 or BMP-4 (inhibitors of caspase 8, a TNF-α activator), quinapril (an inhibitor of angiotensin II, which upregulates TNF-α), interferons such as IL-11 (which modulate TNF-α receptor expression), and aurin-tricarboxylic acid (which inhibits TNF-α), guanidinoethyldisulfide, or a combination thereof.
  • Exemplary anti-inflammatory agents include, for example, naproxen; diclofenac; celecoxib; sulindac; diflunisal; piroxicam; indomethacin; etodolac; meloxicam; ibuprofen; ketoprofen; r-flurbiprofen; mefenamic; nabumetone; tolmetin, and sodium salts of each of the foregoing; ketorolac bromethamine; ketorolac tromethamine; ketorolac acid; choline magnesium trisalicylate; rofecoxib; valdecoxib; lumiracoxib; etoricoxib; aspirin; salicylic acid and its sodium salt; salicylate esters of alpha, beta, gamma-tocopherols and tocotrienols (and all their d, 1, and racemic isomers); methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, t-butyl, esters of acetylsalicylic acid; tenoxicam; aceclofenac; nimesulide; nepafenac; amfenac; bromfenac; flufenamate; phenylbutazone, or a combination thereof.
  • An anti-inflammatory agent can be a steroid. Exemplary steroids include, for example, 21-acetoxypregnenolone, alclometasone, algestone, amcinonide, beclomethasone, betamethasone, budesonide, chloroprednisone, clobetasol, clobetasone, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacort, desonide, desoximetasone, dexamethasone, dexamethasone 21-acetate, dexamethasone 21-phosphate di-Na salt, diflorasone, diflucortolone, difluprednate, enoxolone, fluazacort, fluclorinide, flumethasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, fluorometholone, fluperolone acetate, fluprednidene acetate, fluprednisolone, flurandrenolide, fluticasone propionate, formocortal, halcinonide, halobetasol propionate, halometasone, halopredone acetate, hydrocortamate, hydrocortisone, loteprednol etabonate, mazipredone, medrysone, meprednisone, methylprednisolone, mometasone furoate, paramethasone, prednicarbate, prednisolone, prednisolone 25-diethylamino-acetate, prednisolone sodium phosphate, prednisone, prednival, prednylidene, rimexolone, tixocortol, triamcinolone, triamcinolone acetonide, triamcinolone benetonide, triamcinolone hexacetonide or a combination thereof.
  • Examples of a useful statin for treatment of pain and/or inflammation include, but is not limited to, atorvastatin, simvastatin, pravastatin, cerivastatin, mevastatin (see U.S. Pat. No. 3,883,140, the entire disclosure is herein incorporated by reference), velostatin (also called synvinolin; see U.S. Pat. Nos. 4,448,784 and 4,450,171 these entire disclosures are herein incorporated by reference), fluvastatin, lovastatin, rosuvastatin and fluindostatin (Sandoz XU-62-320), dalvastatin (EP Appln. Publn. No. 738510 A2, the entire disclosure is herein incorporated by reference), eptastatin, pitavastatin, or pharmaceutically acceptable salts thereof or a combination thereof. In various embodiments, the statin may comprise mixtures of (+)R and (−)—S enantiomers of the statin. In various embodiments, the statin may comprise a 1:1 racemic mixture of the statin.
  • Anti-inflammatory agents also include those with anti-inflammatory properties, such as, for example, amitriptyline, carbamazepine, gabapentin, pregabalin, clonidine, or a combination thereof.
  • Unless otherwise specified or apparent from context, where this specification and the set of claims that follows refer to an anti-inflammatory agent, the inventors are also referring to a pharmaceutically acceptable salt of the anti-inflammatory agent including stereoisomers. Pharmaceutically acceptable salts include those salt-forming acids and bases that do not substantially increase the toxicity of the compound. Some examples of potentially suitable salts include salts of alkali metals such as magnesium, calcium, sodium, potassium and ammonium, salts of mineral acids such as hydrochloric, hydriodic, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acids, as well as salts of organic acids such as tartaric, acetic, citric, malic, benzoic, glycollic, gluconic, gulonic, succinic, arylsulfonic, e.g., p-toluenesulfonic acids, or the like.
  • Similarly, when referring to an analgesic agent, unless otherwise specified or apparent from context, it is understood that the inventor is also referring to pharmaceutically acceptable salts including stereoisomers. Pharmaceutically acceptable salts include those salt-forming acids and bases that do not substantially increase the toxicity of the compound. Some examples of potentially suitable salts include salts of alkali metals such as magnesium, calcium, sodium, potassium and ammonium, salts of mineral acids such as hydrochloric, hydriodic, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acids, as well as salts of organic acids such as tartaric, acetic, citric, malic, benzoic, glycollic, gluconic, gulonic, succinic, arylsulfonic, e.g., p-toluenesulfonic acids, or the like.
  • A “drug depot” is the composition in which at least one anti-inflammatory agent and at least one analgesic agent or the pharmaceutically acceptable salts of either or both are administered to the body. Thus, a drug depot may comprise a physical structure to facilitate implantation and retention in a desired site (e.g., nasal cavity, sinus cavity, cardiac site of the patient, particularly at or near a site of surgery, or other site of inflammation, etc.). The drug depot also comprises the drug itself. The term “drug” as used herein is generally meant to refer to any substance that alters the physiology of a patient. The term “drug” may be used interchangeably herein with the terms “therapeutic agent,” “therapeutically effective amount,” and “active pharmaceutical ingredient” or “API.” It will be understood that unless otherwise specified a “drug” formulation may include more than one therapeutic agent, wherein exemplary combinations of therapeutic agents include a combination of two or more drugs. The drug provides a concentration gradient of the therapeutic agent for delivery to the site. In various embodiments, the drug depot provides an optimal drug concentration gradient of the therapeutic agent at a distance of up to about 0.1 cm to about 5 cm from the implant site (e.g., within the nasal and/or sinus cavity or at or near cardiac tissue), and comprises at least one anti-inflammatory agent or its pharmaceutically acceptable salt and/or at least one analgesic agent or its pharmaceutically acceptable salt.
  • A “depot” includes but is not limited to capsules, microspheres, microparticles, microcapsules, microfibers particles, nanospheres, nanoparticles, coating, matrices, wafers, film, strip, ribbon, sponge, patch, pills, pellets, emulsions, liposomes, micelles, gels, or other pharmaceutical delivery compositions or a combination thereof. The drug depot may comprise a pump that holds and administers the pharmaceutical (e.g., anti-inflammatory and/or analgesic). In some embodiments, the drug depot has pores that allow release of the drug from the depot. The drug depot will allow fluid in the depot to displace the drug. However, cell infiltration into the depot will be prevented by the size of the pores of the depot. In this way, in some embodiments, the depot should not function as a tissue scaffold and allow tissue growth. Rather, the drug depot will solely be utilized for drug delivery. In some embodiments, the pores in the drug depot will be less than 250 to 500 microns. This pore size will prevent cells from infiltrating the drug depot and laying down scaffolding cells. Thus, in this embodiment, drug will elute from the drug depot as fluid enters the drug depot, but cells will be prevented from entering. In some embodiments, where there are little or no pores, the drug will elute out from the drug depot by the action of enzymes, by hydrolytic action and/or by other similar mechanisms in the human body.
  • Suitable drug depots for use in the present application are described in U.S. Provisional Application No. 61/046,246 filed Apr. 18, 2008, U.S. Provisional Application No. 61/046,218 filed Apr. 18, 2008, U.S. Provisional Application No. 61/046,218 filed Apr. 18, 2008, U.S. Provisional Application No. 61/046,201 filed Apr. 18, 2008, U.S. Ser. No. 12/105,864 filed Apr. 18, 2008 and U.S. Ser. No. 12/105,375 filed Apr. 18, 2008. The entire disclosure of these applications is herein incorporated by reference into the present application.
  • Suitable materials for the depot are ideally pharmaceutically acceptable biodegradable and/or any bioabsorbable materials that are preferably FDA approved or GRAS materials. These materials can be polymeric or non-polymeric, as well as synthetic or naturally occurring, or a combination thereof. In various embodiments, the drug depot may not be biodegradable or comprise material that is not biodegradable. Non-biodegradable polymers include, but are not limited to, various cellulose derivatives (carboxymethyl cellulose, cellulose acetate, cellulose acetate propionate, ethyl cellulose, hydroxypropyl methyl cellulose, hydroxyalkyl methyl celluloses, and alkyl celluloses), silicon and silicon-based polymers (such as polydimethylsiloxane), polyethylene-co-(vinyl acetate), poloxamer, polyvinylpyrrolidone, poloxamine, polypropylene, polyamide, polyacetal, polyester, poly ethylene-chlorotrifluoroethylene, polytetrafluoroethylene (PTFE or “Teflon™”), styrene butadiene rubber, polyethylene, polypropylene, polyphenylene oxide-polystyrene, poly-α-chloro-p-xylene, polymethylpentene, polysulfone, non-degradable ethylene-vinyl acetate (e.g., ethylene vinyl acetate disks and poly(ethylene-co-vinyl acetate)), and other related biostable polymers or combinations thereof.
  • The drug depot may comprise non-resorbable polymers as well. These non-resorbable polymers can include, but are not limited to, delrin, polyurethane, copolymers of silicone and polyurethane, polyolefins (such as polyisobutylene and polyisoprene), acrylamides (such as polyacrylic acid and poly(acrylonitrile-acrylic acid)), neoprene, nitrile, acrylates (such as polyacrylates, poly(2-hydroxy ethyl methacrylate), methyl methacrylate, 2-hydroxyethyl methacrylate, and copolymers of acrylates with N-vinyl pyrrolidone), N-vinyl lactams, polyacrylonitrile, glucomannan gel, vulcanized rubber and combinations thereof. Examples of polyurethanes include thermoplastic polyurethanes, aliphatic polyurethanes, segmented polyurethanes, hydrophilic polyurethanes, polyether-urethane, polycarbonate-urethane and silicone polyether-urethane. Typically, the non-degradable drug depots may need to be removed.
  • A “therapeutically effective amount” or “effective amount” is such that when administered, the drug results in alteration of the biological activity, such as, for example, inhibition of inflammation, reduction or alleviation of pain, improvement in the condition through the reduction in edema etc. The dosage administered to a patient can unless otherwise specified or apparent from context be as single or multiple doses depending upon a variety of factors, including the drug's administered pharmacokinetic properties, the route of administration, patient conditions and characteristics (sex, age, body weight, health, size, etc.), extent of symptoms, concurrent treatments, frequency of treatment and the effect desired. In some embodiments, because the analgesic and/or anti-inflammatory agent is locally administered, therapeutically effective doses may be less than doses administered by other routes (oral, topical, etc.). For example, the drug dose delivered from the drug depot may be, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or 99.9% less than the oral dosage or injectable dose. In turn, systemic side effects, such as for example, liver transaminase elevations, hepatitis, liver failure, myopathy, constipation, etc. may be reduced or eliminated.
  • In some embodiments the depot is designed for immediate release. In other embodiments the drug depot is designed for sustained release. In other embodiments, the drug depot comprises one or more immediate release surfaces and one or more sustain release surfaces.
  • The phrases “sustained release” or “sustain release” (also referred to as extended release or controlled release) are used herein to refer to one or more therapeutic agent(s) that is introduced into the body of a human or other mammal and continuously or continually releases a stream of one or more therapeutic agents over a predetermined time period and at a therapeutic level sufficient to achieve a desired therapeutic effect throughout the predetermined time period (e.g., certain dose/per day). Reference to a continuous or continual release stream is intended to encompass release that occurs as the result of biodegradation in vivo of the drug depot, or a matrix or component thereof, or as the result of metabolic transformation or dissolution of the therapeutic agent(s) or conjugates of therapeutic agent(s). As persons of ordinary skill are aware, sustained release formulations may, by way of example, be created as films, slabs, strip, fibers, sponges, or gels. In some embodiments, microparticles, microspheres, microcapsules, spheroids, shaped derivatives or pastes are disposed in the film or sponge. The formulations may be in a form that is suitable for suspension in isotonic saline, physiological buffer or other solution acceptable for injection into a patient. Further, the formulations may be used in conjunction with any implantable, insertable or injectable system that a person of ordinary skill would appreciate as useful in connection with embodiments herein including but not limited to parenteral formulations, microspheres, microcapsules, gels, pastes, implantable rods, pellets, films, strips, plates or fibers, etc.
  • The phrase “immediate release” is used herein to refer to one or more therapeutic agent(s) that is introduced into the body and that is allowed to dissolve in or become absorbed at the location to which it is administered, with no intention of delaying or prolonging the dissolution or absorption of the drug. Immediate release refers to the release of drug within a short time period following administration, e.g., generally within a few minutes to about 1 hour.
  • The term “mammal” refers to organisms from the taxonomy class “mammalian,” including but not limited to humans, other primates such as chimpanzees, apes, orangutans and monkeys, rats, mice, cats, dogs, cows, horses, etc. In various embodiments, the mammal is a human patient.
  • The phrase “release rate profile” refers to the percentage of active ingredient that is released over fixed units of time, e.g., mcg/hr, mcg/day, mg/hr, mg/day, 10% per day for ten days, etc. As persons of ordinary skill know, a release rate profile may be but need not be linear. By way of a non-limiting example, the drug depot may be in a film or strip or patch form that releases at least one analgesic agent and at least one anti-inflammatory agent over a period of time.
  • Treating or treatment of a disease or condition (e.g., pain and/or inflammation) refers to executing a protocol, which may include administering one or more drugs to a patient (human, normal or otherwise, or other mammal), in an effort to alleviate signs or symptoms of the disease. Alleviation can occur prior to signs or symptoms of the disease or condition appearing, as well as after their appearance. Thus, “treating” or “treatment” includes “preventing” or “prevention” of disease or undesirable condition (e.g., pain and/or inflammation). In addition, “treating” or “treatment” does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols that have only a marginal effect on the patient. “Reducing pain and/or inflammation” includes a decrease in pain and/or inflammation and does not require complete alleviation of pain and/or inflammation signs or symptoms, and does not require a cure. In various embodiments, reducing pain and/or inflammation includes even a marginal decrease in pain and/or inflammation. By way of example, the administration of the effective dosages of at least one analgesic agent and at least one anti-inflammatory agent may be used to prevent, treat or relieve the symptoms of pain and/or inflammation for different diseases or conditions. These diseases/conditions may comprise chronic inflammatory diseases, including, but not limited to sinusitis, (acute and chronic), rhinitis, nasal and or sinus infection, nasal and/or sinus surgery, epitaxis (nose bleeds), sinus bleeding, nasal and/or sinus obstruction, nasal and/or sinus polyps, nasal and/or sinus cancer, nasal and/or sinus trauma. The drug depot may be used to treat, prevent or reduce diseases/conditions, such as cardiovascular disease. Cardiovascular disease (CVD) is a general term used to classify numerous conditions that affect the heart, heart valves, blood, and vasculature of the body. Cardiovascular diseases include coronary artery disease, angina pectoris, myocardial infarction, atherosclerosis, congestive heart failure, hypertension, cerebrovascular disease, stroke, transient ischemic attacks, cardiomyopathy, arrhythmias, aortic stenosis, or aneurysm.
  • “Localized” delivery includes delivery where one or more drugs are deposited at or near or within a tissue, for example, within the nasal or sinus cavities or cardiac tissue or in close proximity (e.g., within about 5 cm, or preferably within 0.1 cm) thereto. A “targeted delivery system” provides delivery of one or more drugs depots, gels or depot dispersed in the gel having a quantity of therapeutic agent that can be deposited at or near the target site (e.g., nasal or sinus cavity or cardiac tissue) as needed for treatment of pain, inflammation or other disease or condition.
  • The term “biodegradable” includes that all or parts of the drug depot will degrade over time by the action of enzymes, by hydrolytic action and/or by other similar mechanisms in the human body. In various embodiments, “biodegradable” includes that the depot (e.g., strip, film, sheet, etc.) can break down or degrade within the body to non-toxic components after or while a therapeutic agent has been or is being released. By “bioerodible” it is meant that the depot will erode or degrade over time due, at least in part, to contact with substances found in the surrounding tissue, fluids or by cellular action. By “bioabsorbable” it is meant that the depot will be broken down and absorbed within the human body, for example, by a cell or tissue. “Biocompatible” means that the depot will not cause substantial tissue irritation or necrosis at the target tissue site.
  • The phrase “pain management medication” includes one or more therapeutic agents that are administered to prevent, alleviate or remove pain entirely. These therapeutic agents include anti-inflammatory agents, muscle relaxants, analgesics, anesthetics, narcotics, and so forth, and combinations thereof.
  • In various embodiments, the depot (e.g., strip, film, patch, sponge) can be designed to cause an initial burst dose of therapeutic agent within the first 24 hours, 2 days, 3 days, 4 days, or 5 days after implantation. “Initial burst” or “burst effect” or “bolus dose” refer to the release of therapeutic agent from the depot during the first 24 hours, 2 days, 3 days, 4 days, or 5 days after the depot comes in contact with an aqueous fluid (e.g., synovial fluid, cerebral spinal fluid, etc.). This burst effect is particularly beneficial for the analgesic, while in various embodiments, for the anti-inflammatory agent a more linear release of a longer duration may be desired. The “burst effect” is believed to be due to the increased release of therapeutic agent from the depot. In alternative embodiments, the depot (e.g., gel) is designed to avoid this initial burst effect.
  • The drug depot comprising at least one analgesic agent or its pharmaceutically acceptable salt and/or at least one anti-inflammatory agent or its pharmaceutically acceptable salt may be co-administered with a muscle relaxant. Co-administration may involve administering at the same time in separate drug depots or formulating together in the same drug depot.
  • Exemplary muscle relaxants include by way of example and not limitation, alcuronium chloride, atracurium bescylate, baclofen, carboxonium, carisoprodol, chlorphenesin carbamate, chlorzoxazone, cyclobenzaprine, dantrolene, decamethonium bromide, fazadinium, gallamine triethiodide, hexafluorenium, meladrazine, mephensin, metaxalone, methocarbamol, metocurine iodide, pancuronium, pridinol mesylate, styramate, suxamethonium, suxethonium, thiocolchicoside, tizanidine, tolperisone, tubocuarine, vecuronium, or combinations thereof.
  • The drug depot may also comprise other therapeutic agents or active ingredients in addition to the at least one analgesic agent or its pharmaceutically acceptable salt and/or at least one anti-inflammatory agent or its pharmaceutically acceptable salt. Suitable additional therapeutic agents include, but are not limited to, integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, CTLA4-Ig agonists/antagonists (BMS-188667), CD40 ligand antagonists, Humanized anti-IL-6 mAb (MRA, Tocilizumab, Chugai), HMGB-1 mAb (Critical Therapeutics Inc.), anti-IL2R antibodies (daclizumab, basilicimab), ABX (anti IL-8 antibodies), recombinant human IL-10, or HuMax IL-15 (anti-IL 15 antibodies).
  • Other suitable therapeutic agents that may be co-administered with the anti-inflammatory agent and/or analgesic agent include IL-1 inhibitors, such Kineret® (anakinra) which is a recombinant, non-glycosylated form of the human interleukin-1 receptor antagonist (IL-1Ra), or AMG 108, which is a monoclonal antibody that blocks the action of IL-1. Therapeutic agents also include excitatory amino acids such as glutamate and aspartate, antagonists or inhibitors of glutamate binding to NMDA receptors, AMPA receptors, and/or kainate receptors. It is contemplated that where desirable a pegylated form of the above may be used. Examples of other therapeutic agents include NF kappa B inhibitors such as glucocorticoids, antioxidants, such as dilhiocarbamate.
  • Specific examples of additional therapeutic agents suitable for use include, but are not limited to, an anabolic growth factor or anti-catabolic growth factor, analgesic agent, or an osteoinductive growth factor or a combination thereof.
  • Suitable anabolic growth or anti-catabolic growth factors include, but are not limited to, a bone morphogenetic protein, a growth differentiation factor, a LIM mineralization protein, CDMP or progenitor cells or a combination thereof.
  • Suitable analgesic agents include, but are not limited to, acetaminophen, bupivacaine, opioid analgesics such as amitriptyline, carbamazepine, gabapentin, pregabalin, clonidine, opioid analgesics or a combination thereof. Opioid analgesics include, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levorphanol, levophenacylmorphan, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, nalbuphine, normorphine, norpinanone, opium, oxycodone, oxymorphone, papavereturn, pentazocine, phenadoxone, phenomorphan, phanazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propoxyphene, sulfentanil, tilidine, tramadol or a combination thereof.
  • For each anti-inflammatory agent and/or analgesic agent, in some embodiments, the release of each compound may be for at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or at least fifteen days, or longer.
  • The therapeutic agent also includes its pharmaceutically acceptable salt. As used herein, “pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds (e.g., esters or amines) wherein the parent compound may be modified by making acidic or basic salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, or nitric acids; or the salts prepared from organic acids such as acetic, fuoric, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic acid. Pharmaceutically acceptable also includes the racemic mixtures ((+)—R and (−)—S enantiomers) or each of the dextro and levo isomers of the therapeutic agent individually. The therapeutic agent may be in the free acid or base form or be pegylated for long acting activity.
  • Sulfazalazine
  • In one embodiment, the anti-inflammatory agent in the drug depot comprises sulfasalazine. Sulfasalazine is also known as 6-oxo-3-((4-(pyridin-2-ylsulfamoyl)phenyl) hydrazinylidene]cyclohexa-1,4-diene-1-carboxylic acid. Sulfasalazine or a pharmaceutically acceptable salt thereof is available from various pharmaceutical manufacturers. In one embodiment, the dosage of sulfasalazine is from approximately 0.005 μg/day to approximately 3000 mg/day. Additional dosages of sulfasalazine include from approximately 0.005 μg/day to approximately 2000 mg/day; approximately 0.005 μg/day to approximately 1000 mg/day; approximately 0.005 μg/day to approximately 100 mg/day; approximately 0.005 μg/day to approximately 1 mg/day; approximately 0.005 μg/day to approximately 80 μg/day; approximately 0.01 to approximately 70 μg/day; approximately 0.01 to approximately 65 μg/day; approximately 0.01 to approximately 60 μg/day; approximately 0.01 to approximately 55 μg/day; approximately 0.01 to approximately 50 μg/day; approximately 0.01 to approximately 45 μg/day; approximately 0.01 to approximately 40 μg/day; approximately 0.025 to approximately 35 μg/day; approximately 0.025 to approximately 30 μg/day; approximately 0.025 to approximately 25 μg/day; approximately 0.025 to approximately 20 μg/day; and approximately 0.025 to approximately 15 μg/day. In another embodiment, the dosage of sulfasalazine is from approximately 0.05 to approximately 15 μg/day. In another embodiment, the dosage of sulfasalazine is from approximately 0.05 to approximately 10 μg/day.
  • Sulindac
  • In one embodiment, the anti-inflammatory agent in the drug depot comprises sulindac. Sulindac, also known as 2-[6-fluoro-2-methyl-3-[(4-methylsulfinylphenyl)-methylidene]inden-1-yl]-acetic acid may be represented by the formula C20H17FO3S. Sulindac or a pharmaceutically acceptable salt thereof is available from various pharmaceutical manufacturers.
  • The dosage of sulindac may be from approximately 0.001 μg/day to approximately 400 mg/day. Additional dosages of sulindac include from approximately 0.001 μg/day to approximately 200 mg/day; approximately 0.001 μg/day to approximately 100 mg/day; approximately 0.001 μg/day to approximately 1 mg/day; approximately 0.001 to approximately 500 μg/day; approximately 0.001 to approximately 100 μg/day; approximately 0.025 to approximately 75 μg/day; approximately 0.025 to approximately 65 μg/day; approximately 0.025 to approximately 60 μg/day; approximately 0.025 to approximately 55 μg/day; approximately 0.025 to approximately 50 μg/day; approximately 0.025 to approximately 45 μg/day; approximately 0.025 to approximately 40 μg/day; approximately 0.025 to approximately 35 μg/day; approximately 0.005 to approximately 30 μg/day; approximately 0.005 to approximately 25 μg/day; approximately 0.005 to approximately 20 μg/day; and approximately 0.005 to approximately 15 μg/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 15 μg/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 10 μg/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 5 μg/day. In another embodiment, the dosage of sulindac is from approximately 0.01 to approximately 20 μg/day. In another embodiment, the sulindac is administered in a drug depot that releases 9.6 μg/day.
  • Clonidine
  • In one embodiment, the anti-inflammatory agent in the depot is clonidine, also referred to as 2,6-dichloro-N-2-imidazolidinyldenebenzenamine. Clonidine or a pharmaceutically acceptable salt thereof is available from various pharmaceutical manufactures.
  • The dosage may be from approximately 0.0005 to approximately 960 μg/day. Additional dosages of clonidine include from approximately 0.0005 to approximately 900 μg/day; approximately 0.0005 to approximately 500 μg/day; approximately 0.0005 to approximately 250 μg/day; approximately 0.0005 to approximately 100 μg/day; approximately 0.0005 to approximately 75 μg/day; approximately 0.001 to approximately 70 μg/day; approximately 0.001 to approximately 65 μg/day; approximately 0.001 to approximately 60 μg/day; approximately 0.001 to approximately 55 μg/day; approximately 0.001 to approximately 50 μg/day; approximately 0.001 to approximately 45 μg/day; approximately 0.001 to approximately 40 μg/day; approximately 0.001 to approximately 35 μg/day; approximately 0.0025 to approximately 30 μg/day; approximately 0.0025 to approximately 25 μg/day; approximately 0.0025 to approximately 20 μg/day; approximately 0.0025 to approximately 15 μg/day; approximately 0.0025 to approximately 10 μg/day; approximately 0.0025 to approximately 5 μg/day; and approximately 0.0025 to approximately 2.5 μg/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to approximately 15 μg/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to approximately 10 μg/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to approximately 5 μg/day. In another embodiment, the dosage of clonidine is from approximately 0.005 to 2.5 μg/day. In some embodiments, the amount of clonidine is between 40 and 600 μg/day. In some embodiments, the amount of clonidine is between 200 and 400 μg/day.
  • Fluocinolone
  • In one embodiment, the anti-inflammatory agent in the drug depot comprises fluocinolone or a pharmaceutically acceptable salt thereof such as the acetonide salt. Fluocinolone is available from various pharmaceutical manufacturers. The dosage of fluocinolone may be from approximately 0.0005 to approximately 100 μg/day. Additional dosages of fluocinolone include from approximately 0.0005 to approximately 50 μg/day; approximately 0.0005 to approximately 25 μg/day; approximately 0.0005 to approximately 10 μg/day; approximately 0.0005 to approximately 5 μg/day; approximately 0.0005 to approximately 1 μg/day; approximately 0.0005 to approximately 0.75 μg/day; approximately 0.0005 to approximately 0.5 μg/day; approximately 0.0005 to approximately 0.25 μg/day; approximately 0.0005 to approximately 0.1 μg/day; approximately 0.0005 to approximately 0.075 μg/day; approximately 0.0005 to approximately 0.05 μg/day; approximately 0.001 to approximately 0.025 μg/day; approximately 0.001 to approximately 0.01 μg/day; approximately 0.001 to approximately 0.0075 μg/day; approximately 0.001 to approximately 0.005 μg/day; approximately 0.001 to approximately 0.025 μg/day; and approximately 0.002 μg/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to approximately 15 μg/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to approximately 10 μg/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to approximately 5 μg/day. In another embodiment, the dosage of fluocinolone is from approximately 0.001 to 2.5 μg/day. In some embodiments, the amount of fluocinolone is between 40 and 600 μg/day. In some embodiments, the amount of fluocinolone is between 200 and 400 μg/day.
  • Dexamethasone
  • In one embodiment, the anti-inflammatory agent in the drug depot is dexamethasone free base or dexamethasone acetate, also referred to as 8S,9R,10S,11S,13S,14S,16R,17R)-9-Fluoro-11,17-dihydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,11,12,14,15,16 octahydrocyclopenta[a]-phenanthren-3-one), or a pharmaceutically acceptable salt thereof, which is available from various manufacturers.
  • In various embodiments, dexamethasone may be released from the depot at a dose of about 10 pg to about 80 mg/day, about 2.4 ng/day to about 50 mg/day, about 50 ng/day to about 2.5 mg/day, about 250 ng/day to about 250 ug/day, about 250 ng/day to about 50 ug/day, about 250 ng/day to about 25 ug/day, about 250 ng/day to about 1 ug/day, about 300 ng/day to about 750 ng/day or about 0.50 ug/day. In various embodiments, the dose may be about 0.01 to about 10 μg/day or about 1 ng to about 120 μg/day.
  • In one exemplary embodiment, the dexamethasone is dexamethasone sodium phosphate.
  • GED
  • In one embodiment, the therapeutic agent in the drug depot is GED (guanidinoethyldisulfide), which is an inducible nitric oxide synthase inhibitor having anti-inflammatory properties. GED may be in its hydrogen carbonate salt form.
  • The dosage of GED may be from approximately 0.0005 μg/day to approximately 100 mg/day. Additional dosages of GED include from approximately 0.0005 μg/day to approximately 50 mg/day; approximately 0.0005 μg/day to approximately 10 mg/day; approximately 0.0005 μg/day to approximately 1 mg/day; approximately 0.0005 to approximately 800 μg/day; approximately 0.0005 to approximately 50 μg/day; approximately 0.001 to approximately 45 μg/day; approximately 0.001 to approximately 40 μg/day; approximately 0.001 to approximately 35 μg/day; approximately 0.0025 to approximately 30 μg/day; approximately 0.0025 to approximately 25 μg/day; approximately 0.0025 to approximately 20 μg/day; and approximately 0.0025 to approximately 15 μg/day. In another embodiment, the dosage of GED is from approximately 0.005 to approximately 15 μg/day. In another embodiment, the dosage of GED is from approximately 0.005 to approximately 10 μg/day. In another embodiment, the dosage of GED is from approximately 0.005 to approximately 5 μg/day. In another embodiment, the dosage of GED is from approximately 0.005 to 2.5 μg/day. In some embodiments, the amount of GED is between 40 and 600 μg/day. In some embodiments, the amount of GED is between 200 and 400 μg/day.
  • In one exemplary embodiment the dosage of GED is between 0.5 and 4 mg/day. In another exemplary embodiment the dosage of GED is between 0.75 and 3.5 mg/day.
  • Lovastatin
  • In one exemplary embodiment, the anti-inflammatory agent in the depot comprises lovastatin. Lovastatin is a statin that may be obtained from various manufacturers in various forms (e.g., injection, powder, etc.). For example, lovastatin may be obtained from Merck as Mevacor® (see U.S. Pat. No. 4,231,938, the entire disclosure is herein incorporated by reference). Suitable pharmaceutically acceptable salts of lovastatin include one or more compounds derived from bases such as sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, 1-deoxy-2-(methylamino)-D-glucitol, magnesium hydroxide, zinc hydroxide, aluminum hydroxide, ferrous or ferric hydroxide, ammonium hydroxide or organic amines such as N-methylglucamine, choline, arginine or the like or combinations thereof. Suitable pharmaceutically acceptable salts of lovastatin include lithium, calcium, hemicalcium, sodium, potassium, magnesium, aluminum, ferrous or ferric salts thereof or a combination thereof.
  • In various embodiments, the therapeutically effective amount of lovastatin comprises from about 0.1 pg to about 2000 mg, for example, 0.1 ng to 1000 mg, 500 mg, 100 mg, 50 mg, 25 mg, 10 mg, 1 mg, 50 μg, 25 μg, 10 μg, 1 μg, 500 ng, 250 ng, 100 ng, 75 ng, 50 ng, 25 ng, 15 ng, 10 ng, 5 ng, or 1 ng of lovastatin per day. In various embodiments, the dosage may be, for example from about 3 ng/day to 0.3 μg/day.
  • Morphine
  • In one embodiment of the present invention, the analgesic agent in the drug depot is morphine. Morphine is also referred to as (5α,6α)-7,8-didehydro-4,5-epoxy-17-methylmorphinan-3,6-diol and has the chemical formula C17H19NO3. Morphine or a pharmaceutically acceptable salt thereof is available from various manufacturers. In one exemplary embodiment, the morphine comprises morphine sulfate or hydrochloride.
  • The dosage of the morphine may be from 0.1 mg to 1000 mg per day. For example, the dosage of morphine may be for example, 0.1 mg to 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg of morphine per day.
  • Tramadol
  • In one embodiment, the analgesic agent in the drug depot is tramadol. Tramadol is also referred to as (±)cis-2-[(dimethylamino)methyl]-1-(3-methoxyphenyl)cyclohexanol hydrochloride and has the chemical formula C16H25NO2. Tramadol or a pharmaceutically acceptable salt thereof is available from various manufacturers. In various embodiments, tramadol HCL was used.
  • The dosage of the tramadol may be from 0.01 mg to 500 mg per day. For example, the dosage of tramadol may be for example, 0.1 mg to 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, or 500 mg of tramadol per day.
  • In one embodiment, the drug depot contains sufficient tramadol to release between 2.5 and 30 mg/kg/day. In another embodiment the drug depot contains sufficient tramadol to release between 3 and 27.5 mg/kg/day.
  • The at least one anti-inflammatory agent and at least one analgesic agent may also be administered with non-active ingredients. These non-active ingredients may have multi-functional purposes including the carrying, stabilizing and controlling the release of the therapeutic agent(s). The sustained release process, for example, may be by a solution-diffusion mechanism or it may be governed by an erosion-sustained process. Typically, the depot will be a solid or semi-solid formulation comprised of a biocompatible material that can be biodegradable. The term “solid” is intended to mean a rigid material, while “semi-solid” is intended to mean a material that has some degree of flexibility, thereby allowing the depot to bend and conform to the surrounding tissue requirements.
  • In various embodiments, the non-active ingredients will be durable within the tissue site for a period of time equal to (for biodegradable components) or greater than (for non-biodegradable components) the planned period of drug delivery. For example, the depot material may have a melting point or glass transition temperature close to or higher than body temperature, but lower than the decomposition or degradation temperature of the therapeutic agent. However, the pre-determined erosion of the depot material can also be used to provide for slow release of the loaded therapeutic agent(s).
  • In various embodiments, the drug depot may not be biodegradable. For example, the drug depot may comprise polyurethane, polyurea, polyether(amide), PEBA, thermoplastic elastomeric olefin, copolyester, and styrenic thermoplastic elastomer, steel, aluminum, stainless steel, titanium, metal alloys with high non-ferrous metal content and a low relative proportion of iron, carbon fiber, glass fiber, plastics, ceramics or combinations thereof. Typically, these types of drug depots may need to be removed.
  • In some instance, it may be desirable to avoid having to remove the drug depot after use. In those instances, the depot may comprise a biodegradable material. There are numerous materials available for this purpose and having the characteristic of being able to breakdown or disintegrate over a prolonged period of time when positioned at or near the target tissue. As a function of the chemistry of the biodegradable material, the mechanism of the degradation process can be hydrolytical or enzymatical in nature, or both. In various embodiments, the degradation can occur either at the surface (heterogeneous or surface erosion) or uniformly throughout the drug delivery system depot (homogeneous or bulk erosion).
  • In various embodiments, the depot may comprise a bioabsorbable, and/or a biodegradable biopolymer that may provide immediate release, or sustained release of the at least one analgesic agent and at least one anti-inflammatory agent. Examples of suitable sustained release biopolymers include but are not limited to poly (alpha-hydroxy acids), poly (lactide-co-glycolide) (PLGA or PLG), polylactide (PLA), polyglycolide (PG), polyethylene glycol (PEG) conjugates of poly (alpha-hydroxy acids), polyorthoesters, polyaspirins, polyphosphagenes, collagen, starch, pre-gelatinized starch, hyaluronic acid, chitosans, gelatin, alginates, albumin, fibrin, vitamin E analogs, such as alpha tocopheryl acetate, d-alpha tocopheryl succinate, D,L-lactide, or L-lactide, -caprolactone, dextrans, vinylpyrrolidone, polyvinyl alcohol (PVA), PVA-g-PLGA, PEGT-PBT copolymer (polyactive), methacrylates, poly (N-isopropylacrylamide), PEO-PPO-PEO (pluronics), PEO-PPO-PAA copolymers, PLGA-PEO-PLGA, PEG-PLG, PLA-PLGA, poloxamer 407, PEG-PLGA-PEG triblock copolymers, SAIB (sucrose acetate isobutyrate) or combinations thereof. As persons of ordinary skill are aware, mPEG may be used as a plasticizer for PLGA, but other polymers/excipients may be used to achieve the same effect. mPEG imparts malleability to the resulting formulations.
  • Where different combinations of polymers are used (bi, tri (e.g., PLGA-PEO-PLGA) or terpolymers), they may be used in different molar ratios, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In various embodiments, for the long release (e.g. 30 days or longer), the depot comprises 50:50 PLGA to 100 PLA. The molecular weight range is 0.45 to 0.8 dI/g.
  • In various embodiments, the molecular weight of the polymer can be a wide range of values. The average molecular weight of the polymer can be from about 1000 to about 10,000,000; or about 1,000 to about 1,000,000; or about 5,000 to about 500,000; or about 10,000 to about 100,000; or about 20,000 to 50,000.
  • In some embodiments, the at least one biodegradable polymer comprises poly(lactic-co-glycolic acid) (PLA) or poly(orthoester) (POE) or a combination thereof. The poly(lactic-co-glycolic acid) may comprise a mixture of polyglycolide (PGA) and polylactide and in some embodiments, in the mixture, there is more polylactide than polyglycolide. In various other embodiments there is 100% polylactide and 0% polyglycolide; 95% polylactide and 5% polyglycolide; 90% polylactide and 10% polyglycolide; 85% polylactide and 15% polyglycolide; 80% polylactide and 20% polyglycolide; 75% polylactide and 25% polyglycolide; 70% polylactide and 30% polyglycolide; 65% polylactide and 35% polyglycolide; 60% polylactide and 40% polyglycolide; 55% polylactide and 45% polyglycolide; 50% polylactide and 50% polyglycolide; 45% polylactide and 55% polyglycolide; 40% polylactide and 60% polyglycolide; 35% polylactide and 65% polyglycolide; 30% polylactide and 70% polyglycolide; 25% polylactide and 75% polyglycolide; 20% polylactide and 80% polyglycolide; 15% polylactide and 85% polyglycolide; 10% polylactide and 90% polyglycolide; 5% polylactide and 95% polyglycolide; and 0% polylactide and 100% polyglycolide.
  • In various embodiments that comprise both polylactide and polyglycolide; there is at least 95% polylactide; at least 90% polylactide; at least 85% polylactide; at least 80% polylactide; at least 75% polylactide; at least 70% polylactide; at least 65% polylactide; at least 60% polylactide; at least 55%; at least 50% polylactide; at least 45% polylactide; at least 40% polylactide; at least 35% polylactide; at least 30% polylactide; at least 25% polylactide; at least 20% polylactide; at least 15% polylactide; at least 10% polylactide; or at least 5% polylactide; and the remainder of the biopolymer being polyglycolide.
  • In various embodiments, the drug depot comprises poly(lactide-co-glycolide) (PLGA), polylactide (PLA), polyglycolide (PGA), D-lactide, D,L-lactide, L-lactide, D,L-lactide-ε-caprolactone, D,L-lactide-glycolide-ε-caprolactone, glycolide-caprolactone or a combination thereof.
  • As persons of ordinary skill in the art are aware, implantable elastomeric depot compositions having a blend of polymers with different end groups are used the resulting formulation will have a lower burst index and a regulated duration of delivery. For example, one may use polymers with acid (e.g., carboxylic acid) and ester end groups (e.g., lauryl, methyl or ethyl ester end groups).
  • Additionally, by varying the comonomer ratio of the various monomers that form a polymer (e.g., the L/G/CL (CL refers to caprolactone, G refers to glycolic acid and L refers to lactic acid) or G/CL ratio for a given polymer) there will be a resulting depot composition having a regulated burst index and duration of delivery. For example, a depot composition having a polymer with a L/G ratio of 50:50 may have a short duration of delivery ranging from about two days to about one month; a depot composition having a polymer with a L/G ratio of 65:35 may have a duration of delivery of about two months; a depot composition having a polymer with a L/G ratio of 75:25 or L/CL ratio of 75:25 may have a duration of delivery of about three months to about four months; a depot composition having a polymer ratio with a L/G ratio of 85:15 may have a duration of delivery of about five months; a depot composition having a polymer with a L/CL ratio of 25:75 or PLA may have a duration of delivery greater than or equal to six months; a depot composition having a terpolymer of CL/G/L with G greater than 50% and L greater than 10% may have a duration of delivery of about one month and a depot composition having a terpolymer of CL/G/L with G less than 50% and L less than 10% may have a duration months up to six months. In general, increasing the G content relative to the CL content shortens the duration of delivery whereas increasing the CL content relative to the G content lengthens the duration of delivery.
  • In some embodiments, the biodegradable polymer comprises at least 10 wt %, at least 50 wt. %, at least 60 wt. %, at least 70 wt. %, at least 80 wt. %, at least 85 wt. %, at least 90 wt. %, at least 95 wt. %, or at least 99 wt. % of the formulation. In some embodiments, the at least one biodegradable polymer and the analgesic and the anti-inflammatory are the only components of the pharmaceutical formulation.
  • In some embodiments, at least 75% of the particles have a size from about 1 micrometer to about 200 micrometers. In some embodiments, at least 85% of the particles have a size from about 1 micrometer to about 100 micrometers. In some embodiments, at least 95% of the particles have a size from about 5 micrometer to about 30 micrometers. In some embodiments, all of the particles have a size from about 10 micrometer to about 30 micrometers.
  • In some embodiments, at least 75% of the particles have a size from about 5 micrometer to about 20 micrometers. In some embodiments, at least 85% of the particles have a size from about 5 micrometers to about 20 micrometers. In some embodiments, at least 95% of the particles have a size from about 5 micrometer to about 20 micrometers. In some embodiments, all of the particles have a size from about 5 micrometer to about 20 micrometers.
  • The depot may optionally contain inactive materials such as buffering agents and pH adjusting agents such as potassium bicarbonate, potassium carbonate, potassium hydroxide, sodium acetate, sodium borate, sodium bicarbonate, sodium carbonate, sodium hydroxide or sodium phosphate; degradation/release modifiers; drug release adjusting agents; emulsifiers; preservatives such as benzalkonium chloride, chlorobutanol, phenylmercuric acetate and phenylmercuric nitrate, sodium bisulfite, sodium bisulfate, sodium thiosulfate, thimerosal, methylparaben, polyvinyl alcohol and phenylethyl alcohol; solubility adjusting agents; stabilizers; and/or cohesion modifiers. Typically, any such inactive materials will be present within the range of 0-75 wt %, and more typically within the range of 0-30 wt %. If the depot is to be placed at or near cardiac tissue, in various embodiments, the depot may comprise sterile preservative free material.
  • The depot can be different sizes, shapes and configurations. There are several factors that can be taken into consideration in determining the size, shape and configuration of the drug depot. For example, both the size and shape may allow for ease in positioning the drug depot at the target tissue site that is selected as the implantation or injection site. In addition, the shape and size of the system should be selected so as to minimize or prevent the drug depot from moving after implantation or injection. In various embodiments, the drug depot can be shaped like a flat surface such as a disc, film, strip or sheet or the like. Flexibility may be a consideration so as to facilitate placement of the drug depot. In various embodiments, the drug depot can be different sizes, for example, the drug depot may be a length of from about 0.5 mm to 5 mm and have a diameter of from about 0.01 to about 2 mm. In various embodiments, the drug depot may have a layer thickness of from about 0.005 to 1.0 mm, such as, for example, from 0.05 to 0.75 mm.
  • In various embodiments, the drug depot may be in the form of a film, patch or strip and may have a thickness of about 500 microns to about 5,000 microns or in some embodiments about 0.1 mm to about 3 mm or in some embodiments about 0.1 mils to about 60 mils.
  • In various embodiments, when the drug depot comprises a film or strip, it may be placed at the incision site before the site is closed. The film or strip may for example be made of thermoplastic materials. Additionally, specific materials that may be advantageous for use in the film or strip include but are not limited to the compounds identified above as sustained release biopolymers. The drug depot may be formed by mixing the at least one analgesic agent and the at least one anti-inflammatory agent with the polymer.
  • Radiographic markers can be included on the drug depot to permit the user to position the depot accurately into the target site of the patient. These radiographic markers will also permit the user to track movement and degradation of the depot at the site over time. In this embodiment, the user may accurately position the depot in the site using any of the numerous diagnostic imaging procedures. Such diagnostic imaging procedures include, for example, X-ray imaging or fluoroscopy. Examples of such radiographic markers include, but are not limited to, barium, calcium phosphate, and/or metal beads or particles. In various embodiments, the radiographic marker could be a spherical shape or a ring around the depot.
  • Packing
  • The drug depot can be part of a packing material or several drug depots can placed on or around each other until the target area is packed. The packing material, among other things, can act as a hemostatic agent and absorb bodily fluid and/or blood during or after surgery. Packing material includes cellulose based materials, such as cellulose gauze made from cotton or regenerated cellulose fiber, regenerated cellulose sponge, other cellulose fibers or the like can be utilized to absorb body fluids and blood during surgery. The packing material can also contain thrombin, chitin, chitosan, fibrin, amorphous fumed silica, gelatin, corn starch, collagen, polyethylene film, polyvinyl acetate, ethylene-vinyl acetate copolymers, metal foils, laminates of cloth or paper, or a plastic film such as for example, resin-like polyethylene, vinyl acetate homopolymers, and ethylene-vinyl acetate, polypropylene, polyesters, PVC, polysaccharides, hyaluronic acid, xanthan, galactomannan, alginate, non-woven fabrics, or the like.
  • Preferably, the packing material reduces risk of contamination or infection and reduces the risk of interfering with the wound healing process and hinder the prompt recovery of the patient.
  • Typically, packing involves the application of packing material, such as for example, gauze, gel, cotton balls, cotton wedges, sponge, or the like to the target tissue site (e.g., nasal and/or sinus cavity, or cardiac tissue). The drug depot may be placed at or near the target tissue site alone or several drug depots can be packed at the target tissue site. Alternatively, the drug depot can be placed at the target tissue site and be packed in by the packing material (e.g., gauze, gel, cotton balls, cotton wedges, etc.) so that the drug depot remains at or near the target tissue site. In this way the packing material reduces or prevents the drug depot from migrating away from the implant site prior to surgical closure. The drug depot will release the therapeutic agent locally at the site of implantation.
  • The packing material may be coated with a therapeutic agent (e.g., antibiotics, petrolatum, etc.). For nasal packings, the packing material can be placed into the nose one layer at a time, folding one layer on top of the other until the area is completely packed. Often the end of the nose may be taped to keep the packing material in place or to prevent the patient from pulling it out. The packing material may be left at the site or degrade over time or it may be removed within 24-48 hours following surgery. Alternatively the packing material can abut firm tissue so that tape is not needed. For example, turbinates are folds of tissue on the insides of the nose. The folds are sufficiently firm to support packing. A piece of gauze or cotton is wedged with the drug depot (e.g., film, strip, etc.) in between the turbinates where the drug depot will release the therapeutic agent in the adjacent blood vessels.
  • FIG. 1A illustrates a magnified top view of one embodiment of the implantable drug depot in the form of a film or strip 10 that has the analgesic and/or an anti-inflammatory agent 12 disposed on or in the film or strip. As the drug depot degrades over time, the therapeutic agent (e.g., analgesic and/or anti-inflammatory) is released locally to the site of implantation.
  • FIG. 1B illustrates a magnified top view of one embodiment of the implantable drug depot in the form of a multiple films or strips 10 that are stacked shown as 17 one on each other that have the analgesic and/or an anti-inflammatory agent 12 disposed on or in the film or strip. The films or strips are shown stacked together and can be used as packing material at or near a cardiac tissue or within the nasal or sinus cavity.
  • FIG. 2 illustrates a magnified side view of one embodiment of the implantable drug depot 10 in the form of a sponge 19 that has the analgesic and/or an anti-inflammatory agent 12 disposed on or in the sponge. As the drug depot degrades over time, the therapeutic agent (e.g., analgesic and/or anti-inflammatory) is released locally to the site of implantation.
  • FIG. 3 illustrates a partial, side sectional view of a human head illustrating the parts of the nasal cavity 14 with the drug depot in the form of a film or strip 10 on packing material 13 administered thereto. The drug depot is held in place and is packed against the anterior nasal cavity against the nasal mucosa, where the therapeutic agent can be released. The packing material holds the drug depot in position at the target tissue site.
  • FIG. 4 illustrates a partial, front sectional view of a human head illustrating portions of a nasal cavity and sinus cavity with the drug depot in the form of a film or strip 11 administered to the sinus frontilis 16 where the therapeutic agent can be released as the film or strip degrades. Thus, localized delivery of the therapeutic agent can be accomplished locally in the sinus cavity to prevent, treat or reduce inflammation and/or pain locally at the site of implantation. Although the sinus frontilis is shown other areas of the sinus cavity can have the drug depot implanted (e.g., sinus sphenoidalis, cellulae ethmoidalis, the sinus maximilaris 18, etc.).
  • Another area that is ripe with pain and/or inflammation is cardiac tissue. FIG. 5 schematically depicts the heart 20 and the target vessel, which is represented by the descending coronary artery 23 that had a surgical procedure indicated by the suture 24 after myocardial infraction. By implanting a plurality of drug depot films or strips 30 and 32 containing an anti-inflammatory or analgesic agent near the surgical site 24, edema, inflammation and/or pain can be reduced to speed the patient's recovery. Alternatively, the films or strips can be part of packing material that can absorb blood and fluid and degrade over time. The drug depot in the form of a film or strip can be placed in or around cardiac tissue. For example, the drug depot in the form of a film or strip can be placed at or near the left coronary artery 21, circumflex artery 22, aorta 25, and right coronary artery 26 or other areas at or near the heart. The drug depot may be implanted at, near or in cardiac tissue, such as for example, pericardium (e.g., serous pericardium, parietal pericardium, fibrous pericardium, visceral pericardium) myocardium, epicardium, or muscle, connective tissue at or near the heart or like tissue.
  • Gel
  • In various embodiments, the drug depot comprises a gel that can be placed at the target tissue site as a film. In various embodiments, the gel has a pre-dosed viscosity in the range of about 1 to about 500 centipoise (cps), 1 to about 200 cps, or 1 to about 100 cps. After the gel is administered to the target site, the viscosity of the gel will increase and the gel will have a modulus of elasticity (Young's modulus) in the range of about 1×104 to about 6×105 dynes/cm2, or 2×104 to about 5×105 dynes/cm2, or 5×104 to about 5×105 dynes/cm2.
  • In one embodiment, a depot comprises an adherent gel comprising at least one analgesic agent and at least one anti-inflammatory agent that is evenly distributed throughout the gel. The gel may be of any suitable type, as previously indicated, and should be sufficiently viscous so as to prevent the gel from migrating from the targeted delivery site once deployed; the gel should, in effect, “stick” or adhere to the targeted tissue site. The gel may, for example, solidify upon contact with the targeted tissue or after deployment from a targeted delivery system. The targeted delivery system may be, for example, a syringe, a catheter, needle or cannula or any other suitable device. The targeted delivery system may inject the gel into or on the targeted tissue site. The therapeutic agent may be mixed into the gel prior to the gel being deployed at the targeted tissue site. In various embodiments, the gel may be part of a two-component delivery system and when the two components are mixed, a chemical process is activated to form the gel and cause it to stick or to adhere to the target tissue.
  • In various embodiments, a gel is provided that hardens or stiffens after delivery. Typically, hardening gel formulations may have a pre-dosed modulus of elasticity in the range of about 1×104 to about 3×105 dynes/cm2, or 2×104 to about 2×105 dynes/cm2, or 5×104 to about 1×105 dynes/cm2. The post-dosed hardening gels (after delivery) may have a rubbery consistency and have a modulus of elasticity in the range of about 1×104 to about 2×106 dynes/cm2, or 1×105 to about 7×105 dynes/cm2, or 2×105 to about 5×105 dynes/cm2.
  • In various embodiments, for those gel formulations that contain a polymer, the polymer concentration may affect the rate at which the gel hardens (e.g., a gel with a higher concentration of polymer may coagulate more quickly than gels having a lower concentration of polymer). In various embodiments, when the gel hardens, the resulting matrix is solid but is also able to conform to the irregular surface of the tissue (e.g., recesses and/or projections in tissue).
  • The percentage of polymer present in the gel may also affect the viscosity of the polymeric composition. For example, a composition having a higher percentage by weight of polymer is typically thicker and more viscous than a composition having a lower percentage by weight of polymer. A more viscous composition tends to flow more slowly. Therefore, a composition having a lower viscosity may be preferred in some instances.
  • In various embodiments, the molecular weight of the gel can be varied by any one of the many methods known in the art. The choice of method to vary molecular weight is typically determined by the composition of the gel (e.g., polymer versus non-polymer). For example in various embodiments, when the gel comprises one or more polymers, the degree of polymerization can be controlled by varying the amount of polymer initiators (e.g. benzoyl peroxide), organic solvents or activator (e.g. DMPT), crosslinking agents, polymerization agent, and/or reaction time.
  • Suitable gel polymers may be soluble in an organic solvent. The solubility of a polymer in a solvent varies depending on the crystallinity, hydrophobicity, hydrogen-bonding and molecular weight of the polymer. Lower molecular weight polymers will normally dissolve more readily in an organic solvent than high-molecular weight polymers. A polymeric gel, which includes a high molecular weight polymer, tends to coagulate or solidify more quickly than a polymeric composition, which includes a low-molecular weight polymer. Polymeric gel formulations, which include high molecular weight polymers, also tend to have a higher solution viscosity than a polymeric gel, which include a low-molecular weight polymer.
  • When the gel is designed to be a flowable gel, it can vary from low viscosity, similar to that of water, to a high viscosity, similar to that of a paste, depending on the molecular weight and concentration of the polymer used in the gel. The viscosity of the gel can be varied such that the polymeric composition can be applied to a patient's tissues by any convenient technique, for example, by brushing, dripping, injecting, or painting. Different viscosities of the gel will depend on the technique used to apply the composition.
  • In various embodiments, the gel has an inherent viscosity (abbreviated as “I.V.” and units are in deciliters/gram), which is a measure of the gel's molecular weight and degradation time (e.g., a gel with a high inherent viscosity has a higher molecular weight and longer degradation time). Typically, a gel with a high molecular weight provides a stronger film or strip and the film or strip takes more time to degrade. In contrast, a gel with a low molecular weight degrades more quickly and provides a softer film or strip. In various embodiments, the gel has a molecular weight, as shown by the inherent viscosity, from about 0.10 dL/g to about 1.2 dL/g or from about 0.10 dL/g to about 0.40 dL/g.
  • In various embodiments, the gel can have a viscosity of about 300 to about 5,000 centipoise (cp). In other embodiments, the gel can have a viscosity of from about 5 to about 300 cps, from about 10 cps to about 50 cps, from about 15 cps to about 75 cps at room temperature. The gel may optionally have a viscosity enhancing agent such as, for example, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl methylcellulose, carboxymethylcellulose and salts thereof, Carbopol, poly-(hydroxyethylmethacrylate), poly-(methoxyethylmethacrylate), poly(methoxyethoxyethyl methacrylate), polymethylmethacrylate (PMMA), methylmethacrylate (MMA), gelatin, polyvinyl alcohols, propylene glycol, PEG 200, PEG 300, PEG 400, PEG 500, PEG 600, PEG 700, PEG 800, PEG 900, PEG 1000, PEG 1450, PEG 3350, PEG 4500, PEG 8000 or combinations thereof.
  • In various embodiments, when a polymer is employed in the gel, the polymeric composition includes about 10 wt % to about 90 wt % or about 30 wt % to about 60 wt % of the polymer.
  • In various embodiments, the gel is a hydrogel made of high molecular weight biocompatible elastomeric polymers of synthetic or natural origin. A desirable property for the hydrogel to have is the ability to respond rapidly to mechanical stresses, particularly shears and loads, in the human body.
  • Hydrogels obtained from natural sources are particularly appealing because they are more likely to be biodegradable and biocompatible for in vivo applications. Suitable hydrogels include natural hydrogels, such as, for example, gelatin, collagen, silk, elastin, fibrin and polysaccharide-derived polymers like agarose, and chitosan, glucomannan gel, hyaluronic acid, polysaccharides, such as cross-linked carboxyl-containing polysaccharides, or a combination thereof. Synthetic hydrogels include, but are not limited to those formed from polyvinyl alcohol, acrylamides such as polyacrylic acid and poly (acrylonitrile-acrylic acid), polyurethanes, polyethylene glycol (e.g., PEG 3350, PEG 4500, PEG 8000), silicone, polyolefins such as polyisobutylene and polyisoprene, copolymers of silicone and polyurethane, neoprene, nitrile, vulcanized rubber, poly(N-vinyl-2-pyrrolidone), acrylates such as poly(2-hydroxy ethyl methacrylate) and copolymers of acrylates with N-vinyl pyrolidone, N-vinyl lactams, polyacrylonitrile or combinations thereof. The hydrogel materials may further be cross-linked to provide further strength as needed. Examples of different types of polyurethanes include thermoplastic or thermoset polyurethanes, aliphatic or aromatic polyurethanes, polyetherurethane, polycarbonate-urethane or silicone polyether-urethane, or a combination thereof.
  • In various embodiments, rather than directly admixing the therapeutic agents into the gel, microspheres may be dispersed within the gel, the microspheres being loaded with at least one analgesic agent and/or at least one anti-inflammatory agent. In one embodiment, the microspheres provide for a sustained release of the at least one analgesic agent and at least one anti-inflammatory agent. In yet another embodiment, the gel, which is biodegradable, prevents the microspheres from releasing the analgesic agent and/or anti-inflammatory agent; the microspheres thus do not release the analgesic agent and/or anti-inflammatory agent until they have been released from the gel. For example, a gel may be deployed around a target tissue site (e.g., a cardiac tissue). Dispersed within the gel is a plurality of microspheres that encapsulate the desired therapeutic agent. Certain of these microspheres degrade once released from the gel, thus releasing the analgesic agent and/or anti-inflammatory agent. The analgesic agents and/or anti-inflammatory agents may be placed into separate microspheres and then the microspheres combined, or the active ingredients can first be combined and then placed into the microspheres together.
  • Microspheres, much like a fluid, may disperse relatively quickly, depending upon the surrounding tissue type, and hence disperse the at least one analgesic agent and at least one anti-inflammatory agent. In some embodiments, the diameter of the microspheres range from about 10 microns in diameter to about 200 microns in diameter. In some embodiments they range from about 20 to 120 microns in diameters.
  • The present invention also contemplates the use of adherent gels to so constrain dispersal of the therapeutic agent. These gels may be deployed, for example, in the sinus cavity, or cardiac tissue, or in surrounding tissue.
  • Cannulas and Needles
  • It will be appreciated by those with skill in the art that the depot can be administered to the target site using a “cannula” or “needle” that can be a part of a drug delivery device e.g., a syringe, a gun drug delivery device, or any medical device suitable for the application of a drug to a targeted organ or anatomic region. The cannula or needle of the drug depot device is designed to cause minimal physical and psychological trauma to the patient.
  • Cannulas or needles include tubes that may be made from materials, such as for example, polyurethane, polyurea, polyether(amide), PEBA, thermoplastic elastomeric olefin, copolyester, and styrenic thermoplastic elastomer, steel, aluminum, stainless steel, titanium, metal alloys with high non-ferrous metal content and a low relative proportion of iron, carbon fiber, glass fiber, plastics, ceramics or combinations thereof. The cannula or needle may optionally include one or more tapered regions. In various embodiments, the cannula or needle may be beveled. The cannula or needle may also have a tip style vital for accurate treatment of the patient depending on the site for implantation. Examples of tip styles include, for example, Trephine, Cournand, Veress, Huber, Seldinger, Chiba, Francine, Bias, Crawford, deflected tips, Hustead, Lancet, or Tuohey. In various embodiments, the cannula or needle may also be non-coring and have a sheath covering it to avoid unwanted needle sticks.
  • The dimensions of the hollow cannula or needle, among other things, will depend on the site for implantation. The thickness of the cannula or needle will also depend on the site of implantation. In various embodiments, the thickness includes, but is not limited to, from about 0.05 to about 1.655. The gauge of the cannula or needle may be the widest or smallest diameter or a diameter in between for insertion into a human or animal body. The widest diameter is typically about 14 gauge, while the smallest diameter is about 25 gauge. In various embodiments the gauge of the needle or cannula is about 18 to about 22 gauge.
  • In various embodiments, like the drug depot and/or gel, the cannula or needle includes dose radiographic markers that indicate location at or near the site beneath the skin, so that the user may accurately position the depot at or near the site using any of the numerous diagnostic imaging procedures. Such diagnostic imaging procedures include, for example, X-ray imaging or fluoroscopy. Examples of such radiographic markers include, but are not limited to, barium, calcium phosphate, and/or metal beads or particles.
  • In various embodiments, the needle or cannula may include a transparent or translucent portion that can be visualizable by ultrasound, fluoroscopy, x-ray, or other imaging techniques. In such embodiments, the transparent or translucent portion may include a radiopaque material or ultrasound responsive topography that increases the contrast of the needle or cannula relative to the absence of the material or topography.
  • Sterilization
  • The drug depot (e.g., strip, sheet, film, etc.) and/or medical device to administer the drug depot may be sterilizable. In various embodiments, one or more components of the drug depot, and/or medical device to administer the drug depot are sterilized by radiation in a terminal sterilization step in the final packaging. Terminal sterilization of a product provides greater assurance of sterility than from processes such as an aseptic process, which require individual product components to be sterilized separately and the final package assembled in a sterile environment.
  • Typically, in various embodiments, gamma radiation is used in the terminal sterilization step, which involves utilizing ionizing energy from gamma rays that penetrates deeply in the device. Gamma rays are highly effective in killing microorganisms, they leave no residues nor have sufficient energy to impart radioactivity to the device. Gamma rays can be employed when the device is in the package and gamma sterilization does not require high pressures or vacuum conditions, thus, package seals and other components are not stressed. In addition, gamma radiation eliminates the need for permeable packaging materials.
  • In various embodiments, electron beam (e-beam) radiation may be used to sterilize one or more components of the device. E-beam radiation comprises a form of ionizing energy, which is generally characterized by low penetration and high-dose rates. E-beam irradiation is similar to gamma processing in that it alters various chemical and molecular bonds on contact, including the reproductive cells of microorganisms. Beams produced for e-beam sterilization are concentrated, highly-charged streams of electrons generated by the acceleration and conversion of electricity. E-beam sterilization may be used, for example, when the drug depot is included in a gel.
  • Other methods may also be used to sterilize the depot and/or one or more components of the device, including, but not limited to, gas sterilization, such as, for example, with ethylene oxide or steam sterilization.
  • Kits
  • In various embodiments, a kit is provided that may include additional parts along with the drug depot and/or medical device combined together to be used to implant the drug depot (e.g., film, strip, etc.). The kit may include the drug depot device in a first compartment. The second compartment may include a canister holding the drug depot and any other instruments needed for the localized drug delivery. A third compartment may include gloves, drapes, wound dressings, packing material, and other procedural supplies for maintaining sterility of the implanting process, as well as an instruction booklet. A fourth compartment may include additional cannulas and/or needles. A fifth compartment may include the agent for radiographic imaging. Each tool may be separately packaged in a plastic pouch that is radiation sterilized. A cover of the kit may include illustrations of the implanting procedure and a clear plastic cover may be placed over the compartments to maintain sterility.
  • Administration
  • In various embodiments, the analgesic and/or anti-inflammatory may be parenterally administered. The term “parenteral” as used herein refers to modes of administration, which bypass the gastrointestinal tract, and include for example, intravenous, intramuscular, continuous or intermittent infusion, intraperitoneal, intrasternal, subcutaneous, intra-operatively, intrathecally, intradiscally, peridiscally, epidurally, perispinally, intranasally, intraarticular injection or combinations thereof.
  • In various embodiments, a method for delivering a therapeutic agent into a surgery site of a patient is provided. The method comprising inserting a cannula at or near a target tissue site and implanting the drug depot at the target site beneath the skin of the patient and brushing, dripping, spraying, injecting, or painting the gel in the target site to hold or have the drug depot adhere to the target site. In this way unwanted migration of the drug depot away from the target site is reduced or eliminated.
  • In various embodiments, because the analgesic and/or anti-inflammatory agent is locally administered, therapeutically effective doses may be less than doses administered by other routes (oral, topical, etc.). For example, the drug dose delivered from the drug depot may be, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, and 95% less than the oral dosage or injectable dose. In turn, systemic side effects, such as for example, liver transaminase elevations, hepatitis, liver failure, myopathy, constipation, etc. may be reduced or eliminated.
  • In various embodiments, to administer the gel having the drug depot dispersed therein to the desired site, first the cannula or needle can be inserted through the skin and soft tissue down to the target tissue site and the gel administered (e.g., brushed, dripped, injected, or painted, etc.) at or near the target site. In those embodiments where the drug depot is separate from the gel, first the cannula or needle can be inserted through the skin and soft tissue down to the site of injection and one or more base layer(s) of gel can be administered to the target site. Following administration of the one or more base layer(s), the drug depot can be implanted on or in the base layer(s) so that the gel can hold the depot in place or reduce migration. If required a subsequent layer or layers of gel can be applied on the drug depot to surround the depot and further hold it in place. Alternatively, the drug depot may be implanted first and then the gel placed (e.g., brushed, dripped, injected, or painted, etc.) around the drug depot to hold it in place. By using the gel, accurate and precise implantation of a drug depot can be accomplished with minimal physical and psychological trauma to the patient. In some embodiments, the gel may also avoid the need to suture the drug depot to the target site reducing physical and psychological trauma to the patient.
  • The at least one analgesic and/or anti-inflammatory agent formulation may be used to form different pharmaceutical preparations (e.g., drug depot film, sheet, strip, etc.). The pharmaceutical preparations may be formed in an administration with a suitable pharmaceutical carrier that may be solid or liquid, and placed in the appropriate form for parenteral or other administration as desired. As persons of ordinary skill are aware, known carriers include but are not limited to water, gelatin, lactose, starches, stearic acid, magnesium stearate, sicaryl alcohol, talc, vegetable oils, benzyl alcohols, gums, waxes, propylene glycol, polyalkylene glycols and other known carriers.
  • Another embodiment provides a method for treating a mammal suffering from pain and/or inflammation, said method comprising administering a therapeutically effective amount of the analgesic and/or anti-inflammatory agent at a target site beneath the skin at or near the target site.
  • In some embodiments, the therapeutically effective dosage amount (e.g., analgesic and/or anti-inflammatory) and the release rate profile are sufficient to reduce inflammation and/or pain for a period of at least one day, for example, 1-90 days, 1-10 days, 1-3 days, 3-7 days, 3-12 days; 3-14 days, 7-10 days, 7-14 days, 7-21 days, 7-30 days, 7-50 days, 7-90 days, 7-140 days, 14-140 days, 3 days to 150 days, or 3 days to 6 months.
  • In some embodiments the at least one analgesic and/or anti-inflammatory or a portion of the at least one analgesic and/or anti-inflammatory is administered as a bolus dose at the target tissue to provide an immediate release of the analgesic and/or anti-inflammatory.
  • In some embodiments there is a composition useful for the treatment of inflammation comprising an effective amount of at least one analgesic and/or anti-inflammatory that is capable of being locally administered to a target tissue site. By way of example, they may be administered locally to the nasal, sinus, and/or cardiac tissue.
  • In some embodiments, the at least one analgesic and/or anti-inflammatory is administered parenterally, e.g., by injection. In some embodiments, the injection is intra-cardiac, which refers to an injection into the cardiac tissue. An injection may also be into a muscle or other tissue. In other embodiments, the analgesic and/or anti-inflammatory is administered by placement into an open patient cavity during surgery.
  • In some embodiments, the formulation is implantable into a surgical site at the time of surgery. The active ingredients may then be released from the depot via diffusion in a sustained fashion over a period of time, e.g., 3-15 days, 5-10 days or 7-10 days post surgery in order to address pain and inflammation. In some embodiments, the active ingredient may provide longer duration of pain and/or inflammation relief for chronic diseases/conditions with release of one or more drugs up to 6 months or 1 year (e.g., 90, 100, 150, 180 days or longer).
  • In some embodiments, the drug depot may release 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof relative to a total amount of at least one analgesic and/or anti-inflammatory agent loaded in the drug depot over a period of 3 to 12 days, 5 to 10 days or 7 to 10 days after the drug depot is administered to the target tissue site. In some embodiments, the active ingredient may provide longer duration of pain and/or inflammation relief for chronic diseases/conditions as discussed above with release of one or more drugs up to 6 months or 1 year (e.g., 90, 100, 150, 180 days or longer).
  • In various embodiments, an implantable drug depot useful for reducing, preventing or treating pain and/or inflammation is provided in a patient in need of such treatment, the implantable drug depot comprising a therapeutically effective amount of a analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salts thereof, the depot being implantable at a site beneath the skin to reduce, prevent or treat pain and/or inflammation, wherein the drug depot (i) comprises one or more immediate release layer(s) that is capable of releasing about 5% to about 20% of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salts thereof relative to a total amount of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof loaded in the drug depot over a first period of up to 48 hours and (ii) one or more sustain release layer(s) that is capable of releasing about 21% to about 99% of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof relative to a total amount of the analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof loaded in the drug depot over a subsequent period of up to 3 days to 6 months.
  • By way of non-limiting example, the target tissue site may comprise at least one sinus cavity, nasal cavity, cardiac tissue, muscle, ligament, tendon, cartilage, spinal disc, spinal foraminal space near the spinal nerve root, facet or spinal canal. Also by way of example, the inflammation may be associated with orthopedic or spine surgery or a combination thereof. By way of further example, the surgery may be arthroscopic surgery, an excision of a mass, hernia repair, spinal fusion, thoracic, cervical, or lumbar surgery, pelvic surgery or a combination thereof. In some embodiments, the active ingredient may provide longer duration of pain and/or inflammation relief for chronic diseases/conditions as discussed above with release of one or more drugs up to 6 months or 1 year (e.g., 90, 100, 150, 180 days or longer).
  • In some embodiments, the at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof is encapsulated in a plurality of depots comprising microparticles, microspheres, microcapsules, and/or microfibers suspended in a gel.
  • In some embodiments, a method is provided of inhibiting pain and/or inflammation in a patient in need of such treatment, the method comprising delivering one or more biodegradable drug depots comprising a therapeutically effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof to a target tissue site beneath the skin before, during or after surgery, wherein the drug depot releases an effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of 3 days to 6 months.
  • In some embodiments, an implantable drug depot useful for preventing or treating pain and/or inflammation in a patient in need of such treatment is provided, the implantable drug depot comprising a therapeutically effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof, the depot being implantable at a site beneath the skin to prevent or treat inflammation, wherein the drug depot releases an effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of 33 days to 6 months.
  • In some embodiments, an implantable drug depot is provided, wherein the drug depot (i) comprises one or more immediate release layer(s) that releases a bolus dose of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof at a site beneath the skin and (ii) one or more sustain release layer(s) that releases an effective amount of at least one analgesic and/or anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of 3 to 12 days or 5 to 10 days or 7 to 10 days or 3 days to 6 months. By way of example, in the drug depot, the one or more immediate release layer(s) may comprise poly (lactide-co-glycolide) (PLGA) and the one or more sustain release layer(s) may comprise polylactide (PLA).
  • In some embodiments, an implantable drug depot useful for reducing, preventing or treating pain and inflammation in a patient is provided, the implantable drug depot in the form of a film or strip comprising a therapeutically effective amount of an analgesic and an anti-inflammatory agent or pharmaceutically acceptable salts thereof and a polymer; wherein the drug depot is implantable at a site beneath the skin to reduce, prevent or treat pain and inflammation, and the depot is capable of releasing (i) about 5% to about 20% of the analgesic or pharmaceutically acceptable salt thereof relative to a total amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof loaded in the drug depot over a first period of up to 72 hours and (ii) about 21% to about 99% of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof relative to a total amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof loaded in the drug depot over a subsequent period of up to 2 weeks.
  • Method of Making
  • In various embodiments, the drug depot comprising the active ingredients can be made by combining a biocompatible polymer and a therapeutically effective amount of the active ingredients or pharmaceutically acceptable salts thereof and forming the implantable drug depot (e.g., strip, sheet, film) from the combination.
  • Various techniques are available for forming at least a portion of a drug depot from the biocompatible polymer(s), therapeutic agent(s), and optional materials, including solution processing techniques and/or thermoplastic processing techniques. Where solution processing techniques are used, a solvent system is typically selected that contains one or more solvent species. The solvent system is generally a good solvent for at least one component of interest, for example, biocompatible polymer and/or therapeutic agent. The particular solvent species that make up the solvent system can also be selected based on other characteristics, including drying rate and surface tension.
  • Solution processing techniques include solvent casting techniques, spin coating techniques, web coating techniques, solvent spraying techniques, dipping techniques, techniques involving coating via mechanical suspension, including air suspension (e.g., fluidized coating), ink jet techniques and electrostatic techniques. Where appropriate, techniques such as those listed above can be repeated or combined to build up the depot to obtain the desired release rate and desired thickness.
  • In various embodiments, a solution containing solvent and biocompatible polymer are combined and placed in a mold of the desired size and shape. In this way, polymeric regions, including barrier layers, therapeutic layers, and so forth can be formed. If desired, the solution can further comprise, one or more of the following: other therapeutic agent(s) and other optional additives such as radiographic agent(s), etc. in dissolved or dispersed form. This results in a polymeric matrix region containing these species after solvent removal. In other embodiments, a solution containing solvent with dissolved or dispersed therapeutic agent is applied to a pre-existing polymeric region, which can be formed using a variety of techniques including solution processing and thermoplastic processing techniques, whereupon the therapeutic agent is imbibed into the polymeric region.
  • Thermoplastic processing techniques for forming the depot or portions thereof include molding techniques (for example, injection molding, rotational molding, and so forth), extrusion techniques (for example, extrusion, co-extrusion, multi-layer extrusion, and so forth) and casting.
  • Thermoplastic processing in accordance with various embodiments comprises mixing or compounding, in one or more stages, the biocompatible polymer(s) and one or more of the following: the active ingredients, optional additional therapeutic agent(s), radiographic agent(s), and so forth. The resulting mixture is then shaped into an implantable drug depot. The mixing and shaping operations may be performed using any of the conventional devices known in the art for such purposes.
  • During thermoplastic processing, there exists the potential for the therapeutic agent(s) to degrade, for example, due to elevated temperatures and/or mechanical shear that are associated with such processing. For example, certain therapeutic agents may undergo substantial degradation under ordinary thermoplastic processing conditions. Hence, processing is preferably performed under modified conditions, which prevent the substantial degradation of the therapeutic agent(s). Although it is understood that some degradation may be unavoidable during thermoplastic processing, degradation is generally limited to 10% or less. Among the processing conditions that may be controlled during processing to avoid substantial degradation of the therapeutic agent(s) are temperature, applied shear rate, applied shear stress, residence time of the mixture containing the therapeutic agent, and the technique by which the polymeric material and the therapeutic agent(s) are mixed.
  • Mixing or compounding biocompatible polymer with therapeutic agent(s) and any additional additives to form a substantially homogenous mixture thereof may be performed with any device known in the art and conventionally used for mixing polymeric materials with additives.
  • Where thermoplastic materials are employed, a polymer melt may be formed by heating the biocompatible polymer, which can be mixed with various additives (e.g., therapeutic agent(s), inactive ingredients, etc.) to form a mixture. A common way of doing so is to apply mechanical shear to a mixture of the biocompatible polymer(s) and additive(s). Devices in which the biocompatible polymer(s) and additive(s) may be mixed in this fashion include devices such as single screw extruders, twin screw extruders, banbury mixers, high-speed mixers, ross kettles, and so forth.
  • Any of the biocompatible polymer(s) and various additives may be premixed prior to a final thermoplastic mixing and shaping process, if desired (e.g., to prevent substantial degradation of the therapeutic agent among other reasons).
  • For example, in various embodiments, a biocompatible polymer is precompounded with a radiographic agent (e.g., radio-opacifying agent) under conditions of temperature and mechanical shear that would result in substantial degradation of the therapeutic agent, if it were present. This precompounded material is then mixed with therapeutic agent under conditions of lower temperature and mechanical shear, and the resulting mixture is shaped into the active ingredient containing drug depot. Conversely, in another embodiment, the biocompatible polymer can be precompounded with the therapeutic agent under conditions of reduced temperature and mechanical shear. This precompounded material is then mixed with, for example, a radio-opacifying agent, also under conditions of reduced temperature and mechanical shear, and the resulting mixture is shaped into the drug depot.
  • The conditions used to achieve a mixture of the biocompatible polymer and therapeutic agent and other additives will depend on a number of factors including, for example, the specific biocompatible polymer(s) and additive(s) used, as well as the type of mixing device used.
  • As an example, different biocompatible polymers will typically soften to facilitate mixing at different temperatures. For instance, where a depot is formed comprising PLGA or PLA polymer, a radio-opacifying agent (e.g., bismuth subcarbonate), and a therapeutic agent prone to degradation by heat and/or mechanical shear (e.g., clonidine), in various embodiments, the PGLA or PLA can be premixed with the radio-opacifying agent at temperatures of about, for example, 150° C. to 170° C. The therapeutic agent is then combined with the premixed composition and subjected to further thermoplastic processing at conditions of temperature and mechanical shear that are substantially lower than is typical for PGLA or PLA compositions. For example, where extruders are used, barrel temperature, volumetric output are typically controlled to limit the shear and therefore to prevent substantial degradation of the therapeutic agent(s). For instance, the therapeutic agent and premixed composition can be mixed/compounded using a twin screw extruder at substantially lower temperatures (e.g., 100-105° C.), and using substantially reduced volumetric output (e.g., less than 30% of full capacity, which generally corresponds to a volumetric output of less than 200 cc/min). It is noted that this processing temperature is well below the melting points of certain active ingredients, such as an anti-inflammatory and analgesic because processing at or above these temperatures will result in substantial therapeutic agent degradation. It is further noted that in certain embodiments, the processing temperature will be below the melting point of all bioactive compounds within the composition, including the therapeutic agent. After compounding, the resulting depot is shaped into the desired form, also under conditions of reduced temperature and shear.
  • In other embodiments, biodegradable polymer(s) and one or more therapeutic agents are premixed using non-thermoplastic techniques. For example, the biocompatible polymer can be dissolved in a solvent system containing one or more solvent species. Any desired agents (for example, a radio-opacifying agent, a therapeutic agent, or both radio-opacifying agent and therapeutic agent) can also be dissolved or dispersed in the solvents system. Solvent is then removed from the resulting solution/dispersion, forming a solid material. The resulting solid material can then be granulated for further thermoplastic processing (for example, extrusion) if desired.
  • As another example, the therapeutic agent can be dissolved or dispersed in a solvent system, which is then applied to a pre-existing drug depot (the pre-existing drug depot can be formed using a variety of techniques including solution and thermoplastic processing techniques, and it can comprise a variety of additives including a radio-opacifying agent and/or viscosity enhancing agent), whereupon the therapeutic agent is imbibed on or in the drug depot. As above, the resulting solid material can then be granulated for further processing, if desired.
  • Typically, an extrusion processes may be used to form the drug depot comprising a biocompatible polymer(s), therapeutic agent(s) and radio-opacifying agent(s). Co-extrusion may also be employed, which is a shaping process that can be used to produce a drug depot comprising the same or different layers or regions (for example, a structure comprising one or more polymeric matrix layers or regions that have permeability to fluids to allow immediate and/or sustained drug release). Multi-region depots can also be formed by other processing and shaping techniques such as co-injection or sequential injection molding technology.
  • In various embodiments, the depot that may emerge from the thermoplastic processing (e.g., film, strip, etc.) is cooled. Examples of cooling processes include air cooling and/or immersion in a cooling bath. In some embodiments, a water bath is used to cool the extruded depot. However, where a water-soluble therapeutic agent such as active ingredients are used, the immersion time should be held to a minimum to avoid unnecessary loss of therapeutic agent into the bath.
  • In various embodiments, immediate removal of water or moisture by use of ambient or warm air jets after exiting the bath will also prevent re-crystallization of the drug on the depot surface, thus controlling or minimizing a high drug dose “initial burst” or “bolus dose” upon implantation or insertion if this is release profile is not desired.
  • In various embodiments, the drug depot can be prepared by mixing or spraying the drug with the polymer and then molding the depot to the desired shape. In various embodiments, active ingredients are used and mixed or sprayed with the PLGA or PEG550 polymer, and the resulting depot may be formed by extrusion and dried.
  • In some embodiments, when the drug depot comprises a film or strip, it may be formed into a film or strip by methods such as extrusion, coating, spreading, casting or the like. If a multi-layered film or strip is desired, this may be accomplished by co-extruding more than one combination of components, which may be of the same or different composition. A multi-layered film or strip may also be achieved by coating, spreading, or casting a combination onto an already formed film layer.
  • Coating or casting methods are particularly useful for the purpose of forming the films or strips. Specific examples include reverse roll coating, gravure coating, immersion or dip coating, metering rod or meyer bar coating, slot die or extrusion coating, gap or knife over roll coating, air knife coating, curtain coating, or combinations thereof, especially when a multi-layered films or strips are desired.
  • It will be apparent to those skilled in the art that various modifications and variations can be made to various embodiments described herein without departing from the spirit or scope of the teachings herein. Thus, it is intended that various embodiments cover other modifications and variations of various embodiments within the scope of the present teachings.

Claims (20)

1. An implantable drug depot useful for reducing, preventing or treating pain and inflammation in a patient in need of such treatment, the implantable drug depot being in the form of a biodegradable film, patch, strip, sheet or sponge and comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent, the depot being implantable at or near a cardiac tissue or within a nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is capable of releasing an effective amount of the analgesic and/or an anti-inflammatory agent over a period of at least one day.
2. An implantable drug depot according to claim 1, wherein the drug depot releases the analgesic and an anti-inflammatory over a period of at least 3 days.
3. An implantable drug depot according to claim 1, wherein the analgesic comprises alfentanil, butorphanol, codeine, fentanyl, hydromorphone, levorphanol, meperidine, morphine, sulfentanil, tramadol or a combination thereof.
4. An implantable drug depot according to claim 1, wherein the anti-inflammatory agent comprises clonidine, fluocinolone, dexamethasone, sulindac, sulfasalazine or a combination thereof.
5. An implantable drug depot according to claim 1, wherein the biodegradable sheet comprises a polymer and the polymer comprises poly(lactide-co-glycolide) (PLGA), polylactide (PLA), polyglycolide (PGA), D-lactide, D,L-lactide, L-lactide, D,L-lactide-ε-caprolactone, D,L-lactide-glycolide-ε-caprolactone or a combination thereof.
6. An implantable drug depot according to claim 1, wherein the drug depot comprises a polymer and the polymer comprises about 60% to 99% of the total weight % of the drug depot.
7. An implantable drug depot according to claim 1, wherein the drug depot releases (i) a bolus dose of the analgesic or pharmaceutically acceptable salt thereof at or near cardiac tissue or within the nasal or sinus cavity over a period of up to 3 days and (ii) an effective amount of the anti-inflammatory agent or pharmaceutically acceptable salt thereof over a period of at least 7 days.
8. An implantable drug depot according to claim 1, wherein the drug depot releases about 20% to about 99% of the analgesic and the anti-inflammatory relative to a total amount of the analgesic and the anti-inflammatory agent loaded in the drug depot over a period of 3 days to 2 weeks after the drug depot is administered at or near a cardiac tissue or within the nasal or sinus cavity.
9. An implantable drug depot according to claim 1, wherein the analgesic and the anti-inflammatory agent are encapsulated in a plurality of depots comprising microparticles, microspheres, microcapsules, and/or microfibers suspended in a film, strip or sponge.
10. An implantable drug depot according to claim 1, wherein the drug depot is in the form of a nasal or cardiac packing film or cardiac patch.
11. A method of making an implantable drug depot of claim 1, the method comprising combining a biocompatible polymer and a therapeutically effective amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof and forming the implantable drug depot from the combination.
12. A method of treating or preventing pain and inflammation in a patient in need of such treatment, the method comprising administering one or more biodegradable drug depots comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent at or near a cardiac tissue or within a nasal or sinus cavity to reduce, prevent or treat pain and/or inflammation, wherein the drug depot is in the form of a biodegradable film, strip or patch that releases an effective amount of the analgesic and/or the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of at least 1 day.
13. A method according to claim 12, wherein the analgesic comprises alfentanil, butorphanol, codeine, fentanyl, hydromorphone, levorphanol, meperidine, morphine, sulfentanil, tramadol or a combination thereof, and the anti-inflammatory agent comprises clonidine, fluocinolone, dexamethasone, sulindac, sulfasalazine or a combination thereof.
14. A method according to claim 12, wherein the drug depot releases 0.1 mg to 100 mg of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof every 24 to 48 hours to reduce, treat or prevent pain and inflammation over a period of 3 days to 2 weeks after the drug depot is administered at or near the cardiac tissue or within the nasal or sinus cavity target tissue site.
15. A method according to claim 12, wherein the drug depot comprises a polymer comprising poly(lactide-co-glycolide) (PLGA), polylactide (PLA), polyglycolide (PGA), D-lactide, D,L-lactide, L-lactide, D,L-lactide-ε-caprolactone, poly(orthoester) (POE), D,L-lactide-glycolide-ε-caprolactone or a combination thereof.
16. A method according to claim 12, wherein the drug depot comprises a polymer and the polymer comprises about 70% to about 90% of the total weight % of the drug depot.
17. A method according to claim 12, wherein the drug depot releases (i) a bolus dose of the analgesic or pharmaceutically acceptable salt thereof at or near cardiac tissue or within the nasal or sinus cavity over a period up to 3 days and (ii) an effective amount of the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of up to 2 weeks.
18. A method of reducing pain and inflammation in a patient in need of such treatment, the method comprising delivering one or more biodegradable drug depots in the form of a biodegradable film or patch comprising a therapeutically effective amount of an analgesic and/or an anti-inflammatory agent or pharmaceutically acceptable salts thereof at or near a cardiac tissue or within a nasal or sinus cavity of the patient, wherein the drug depot releases an effective amount of the analgesic and the anti-inflammatory agent or pharmaceutically acceptable salts thereof over a period of at least 1 day.
19. A method according to claim 18, wherein the drug depot is in the form of a nasal packing film.
20. A method according to claim 18, wherein the drug depot is in the form of a cardiac packing film or patch.
US12/409,058 2009-03-23 2009-03-23 Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue Abandoned US20100239632A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US12/409,058 US20100239632A1 (en) 2009-03-23 2009-03-23 Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
PCT/US2010/028140 WO2010111178A2 (en) 2009-03-23 2010-03-22 Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
EP10756666A EP2410998A4 (en) 2009-03-23 2010-03-22 Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
US15/280,381 US20170014337A1 (en) 2009-03-23 2016-09-29 Drug depots for treatment of pain and inflammation in sinus and nasal cavaties
US16/223,571 US10653619B2 (en) 2009-03-23 2018-12-18 Drug depots for treatment of pain and inflammation

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US12/409,058 US20100239632A1 (en) 2009-03-23 2009-03-23 Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/280,381 Division US20170014337A1 (en) 2009-03-23 2016-09-29 Drug depots for treatment of pain and inflammation in sinus and nasal cavaties

Publications (1)

Publication Number Publication Date
US20100239632A1 true US20100239632A1 (en) 2010-09-23

Family

ID=42737857

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/409,058 Abandoned US20100239632A1 (en) 2009-03-23 2009-03-23 Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
US15/280,381 Abandoned US20170014337A1 (en) 2009-03-23 2016-09-29 Drug depots for treatment of pain and inflammation in sinus and nasal cavaties
US16/223,571 Active US10653619B2 (en) 2009-03-23 2018-12-18 Drug depots for treatment of pain and inflammation

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/280,381 Abandoned US20170014337A1 (en) 2009-03-23 2016-09-29 Drug depots for treatment of pain and inflammation in sinus and nasal cavaties
US16/223,571 Active US10653619B2 (en) 2009-03-23 2018-12-18 Drug depots for treatment of pain and inflammation

Country Status (3)

Country Link
US (3) US20100239632A1 (en)
EP (1) EP2410998A4 (en)
WO (1) WO2010111178A2 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120095418A1 (en) * 2010-10-19 2012-04-19 Joshua Stopek Self-Supporting Films For Delivery Of Therapeutic Agents
JP2012087121A (en) * 2010-10-19 2012-05-10 Tyco Healthcare Group Lp Method of forming self-supporting film for delivery of therapeutic agent
WO2012087606A1 (en) * 2010-12-20 2012-06-28 Saint Joseph's Translational Research Institute, Inc. A drug eluting patch for the treatment of localized tissue disease or defect
WO2012148570A1 (en) * 2011-04-25 2012-11-01 Warsaw Orthopedic, Inc. Medical devices and methods comprising an anabolic agent for wound healing
WO2012166539A1 (en) * 2011-05-31 2012-12-06 Cormatrix Cardiovascular, Inc. Compositions for preventing cardiac arrhythmia
US8449607B2 (en) 2007-12-18 2013-05-28 Cormatrix Cardiovascular, Inc. Prosthetic tissue valve
US8679176B2 (en) 2007-12-18 2014-03-25 Cormatrix Cardiovascular, Inc Prosthetic tissue valve
US8696744B2 (en) 2011-05-27 2014-04-15 Cormatrix Cardiovascular, Inc. Extracellular matrix material valve conduit and methods of making thereof
US20140221447A1 (en) * 2009-10-26 2014-08-07 Warsaw Orthopedic, Inc. Formulation for preventing or reducing bleeding at a surgical site
US20160008286A1 (en) * 2012-02-21 2016-01-14 Auburn University Buprenorphine nanoparticle composition and methods thereof
US20160354992A1 (en) * 2015-06-02 2016-12-08 Biotronik Se & Co. Kg Therapeutic Or Diagnostic Medical Product Having An Adhesion-Enhancing Surface Structure
US9592243B2 (en) 2011-04-25 2017-03-14 Warsaw Orthopedic, Inc. Medical devices and methods comprising an anabolic agent for treatment of an injury
US20180207180A1 (en) * 2017-01-20 2018-07-26 Aurin Biotech Inc. Methods for treating and preventing nociceptive pain
JP2019500944A (en) * 2015-12-08 2019-01-17 メドトロニック・ゾーメド・インコーポレーテッド Dissolvable nasal sinus sponge
WO2019060549A1 (en) * 2017-09-20 2019-03-28 Sinus Clear Innovations, Llc Wound healing device and methods for wound healing
US10596330B2 (en) 2015-08-26 2020-03-24 Medtronic Xomed, Inc. Resorbable, drug-eluting submucosal turbinate implant device and method
US10653619B2 (en) 2009-03-23 2020-05-19 Medtronic, Inc. Drug depots for treatment of pain and inflammation
USRE48948E1 (en) 2008-04-18 2022-03-01 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable polymer

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3691618A1 (en) 2017-10-06 2020-08-12 Foundry Therapeutics, Inc. Implantable depots for the controlled release of therapeutic agents
CA3086446A1 (en) 2017-12-20 2019-06-27 Stryker European Holdings I, Llc Biomedical foam
RU2680244C1 (en) * 2017-12-28 2019-02-19 Общество С Ограниченной Ответственностью "Валента-Интеллект" Combination of flupirtine and cyclobenzaprine for treatment of pain syndromes

Citations (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3020660A (en) * 1959-11-30 1962-02-13 Scherotto John Collapsible imitation tree
US3190802A (en) * 1961-10-09 1965-06-22 Boehringer Sohn Ingelheim Shaving composition and method of using same
US4765974A (en) * 1985-12-24 1988-08-23 Nitto Electric Industrial Co., Ltd. Preparation for percutaneous administration
US5175052A (en) * 1988-05-11 1992-12-29 Nitto Denko Corporation Adhesive tape preparation of clonidine
US5447947A (en) * 1990-02-26 1995-09-05 Arc 1 Compositions and methods of treatment of sympathetically maintained pain
US5484607A (en) * 1993-10-13 1996-01-16 Horacek; H. Joseph Extended release clonidine formulation
US5635204A (en) * 1994-03-04 1997-06-03 Montefiore Medical Center Method for transdermal induction of anesthesia, analgesia or sedation
US5801188A (en) * 1997-01-08 1998-09-01 Medtronic Inc. Clonidine therapy enhancement
US5900245A (en) * 1996-03-22 1999-05-04 Focal, Inc. Compliant tissue sealants
US5942503A (en) * 1995-11-14 1999-08-24 Boehringer Indelheim Kg Use of Epinastine for the treatment of pain
US5942530A (en) * 1997-08-28 1999-08-24 Eli Lilly And Company Method for treating pain
US5945416A (en) * 1996-03-25 1999-08-31 Eli Lilly And Company Method for treating pain
US5980927A (en) * 1995-02-10 1999-11-09 Medtronic, Inc. Method and apparatus for administering analgesics, and method for making same device
US6069129A (en) * 1998-03-13 2000-05-30 Mrs, Llc Elastin derived composition and method of using same
US6147102A (en) * 1999-10-26 2000-11-14 Curatek Pharmaceuticals Holding, Inc. Clonidine preparations
US6179862B1 (en) * 1998-08-14 2001-01-30 Incept Llc Methods and apparatus for in situ formation of hydrogels
US6277413B1 (en) * 1998-07-17 2001-08-21 Skyepharma, Inc. Biodegradable compositions for the controlled release of encapsulated substances
US20010018075A1 (en) * 1997-07-15 2001-08-30 Takada Shigeyuki Process for producing sustained-release preparation
US6287588B1 (en) * 1999-04-29 2001-09-11 Macromed, Inc. Agent delivering system comprised of microparticle and biodegradable gel with an improved releasing profile and methods of use thereof
US6306424B1 (en) * 1999-06-30 2001-10-23 Ethicon, Inc. Foam composite for the repair or regeneration of tissue
US6331311B1 (en) * 1996-12-20 2001-12-18 Alza Corporation Injectable depot gel composition and method of preparing the composition
US20020009454A1 (en) * 1997-02-10 2002-01-24 Amgen Inc. Composition and method for treating inflammatory diseases
US20020058656A1 (en) * 2000-09-19 2002-05-16 Ockert David M. Triple drug therapy for the treatment and prevention of acute or chronic pain
US20020064547A1 (en) * 1998-03-19 2002-05-30 Rey T. Chern Liquid polymeric compositions for controlled release of bioactive substances
US20020090398A1 (en) * 1999-11-16 2002-07-11 Atrix Laboratories, Inc. Biodegradable polymer composition
US20020094998A1 (en) * 2000-11-01 2002-07-18 Burke James A. Methods and compositions for treatment of ocular neovascularization and neural injury
US6428804B1 (en) * 1997-10-27 2002-08-06 Ssp Co., Ltd. Intra-articular preparation for the treatment of arthropathy
US6451335B1 (en) * 1998-07-02 2002-09-17 Euro-Celtique S.A. Formulations and methods for providing prolonged local anesthesia
US20030009145A1 (en) * 2001-03-23 2003-01-09 Struijker-Boudier Harry A.J. Delivery of drugs from sustained release devices implanted in myocardial tissue or in the pericardial space
US20030022926A1 (en) * 2001-05-07 2003-01-30 Lavand'homme Patricia Method for treating neuropathic pain and pharmaceutical preparation therefor
US6589549B2 (en) * 2000-04-27 2003-07-08 Macromed, Incorporated Bioactive agent delivering system comprised of microparticles within a biodegradable to improve release profiles
US20030185872A1 (en) * 2002-03-27 2003-10-02 Frank Kochinke Methods and drug delivery systems for the treatment of orofacial diseases
US6630155B1 (en) * 1998-10-28 2003-10-07 Atrix Laboratories, Inc. Controlled release liquid delivery compositions with low initial drug burst
US6632457B1 (en) * 1998-08-14 2003-10-14 Incept Llc Composite hydrogel drug delivery systems
US20040028726A1 (en) * 1999-11-29 2004-02-12 Wilfried Fischer Transdermal systems for the delivery of clonidine
US20040072799A1 (en) * 2002-07-19 2004-04-15 Omeros Corporation Biodegradable triblock copolymers, synthesis methods therefore, and hydrogels and biomaterials made there from
US20040082540A1 (en) * 2001-11-13 2004-04-29 Hermida Ochoa Elias Humberto Use of a mixture of sodium hyaluronate and chondroitin sulfate for the treatment of osteoarthritis
US20040101582A1 (en) * 2002-11-25 2004-05-27 Richard Wolicki Treatment of neuropathy
US6773714B2 (en) * 1998-10-28 2004-08-10 Atrix Laboratories, Inc. Polymeric delivery formulations of leuprolide with improved efficacy
US20040208917A1 (en) * 2003-04-16 2004-10-21 Wilfried Fischer Transdermal systems for the release of clonidine
US20040265364A1 (en) * 2003-06-25 2004-12-30 Binnur Ozturk Neuropathy cream
US20050043706A1 (en) * 2003-03-14 2005-02-24 Eaton Donald J. Sinus delivery of sustained release therapeutics
US20050059744A1 (en) * 2003-09-12 2005-03-17 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
US20050058696A1 (en) * 2003-09-12 2005-03-17 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
US20050095277A1 (en) * 2003-06-25 2005-05-05 Binnur Ozturk Neuropathy cream
US20050142163A1 (en) * 2003-11-10 2005-06-30 Angiotech International Ag Medical implants and fibrosis-inducing agents
US20050186261A1 (en) * 2004-01-30 2005-08-25 Angiotech International Ag Compositions and methods for treating contracture
US20050197293A1 (en) * 2002-10-28 2005-09-08 Scott Mellis Use of an IL-1 antagonist for treating arthritis
US20050245905A1 (en) * 2004-04-30 2005-11-03 Schmidt Steven P Local drug-delivery system
US20050288620A1 (en) * 2004-06-28 2005-12-29 Shippert Ronald D Nose pack method and apparatus
US6992110B2 (en) * 2001-11-05 2006-01-31 Cypress Bioscience, Inc. Methods of treating fibromyalgia syndrome, chronic fatigue syndrome and pain
US20060148903A1 (en) * 2004-11-24 2006-07-06 Algorx Pharmaceuticals, Inc. Capsaicinoid gel formulation and uses thereof
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060189944A1 (en) * 2005-02-08 2006-08-24 Campbell Patrick K Spray for fluent materials
US20070005094A1 (en) * 2005-04-04 2007-01-04 Eaton Donald J Device and methods for treating paranasal sinus conditions
US20070021358A1 (en) * 2005-05-25 2007-01-25 Elazer Edelman Localized delivery of cardiac inotropic agents
US20070066864A1 (en) * 2005-09-06 2007-03-22 Sean Forde Devices and methods for treating cardiac tissue
US20070104769A1 (en) * 2005-11-04 2007-05-10 Lifescience Plus, Inc. Bioabsorbable hemostatic gauze
US20070156180A1 (en) * 2005-12-30 2007-07-05 Jaax Kristen N Methods and systems for treating osteoarthritis
US20070178138A1 (en) * 2006-02-01 2007-08-02 Allergan, Inc. Biodegradable non-opthalmic implants and related methods
US20070202074A1 (en) * 2003-01-15 2007-08-30 Shalaby Shalaby W Polymeric precursors of non-absorbable, in situ-forming hydrogels and applications thereof
US20070243228A1 (en) * 2006-04-13 2007-10-18 Mckay William F Drug depot implant designs and methods of implantation
US20080009830A1 (en) * 2006-06-27 2008-01-10 Fujimoto Kazuro L Biodegradable elastomeric patch for treating cardiac or cardiovascular conditions
US20080038351A1 (en) * 2006-08-14 2008-02-14 Neal Beals Flowable carrier matrix
US7345065B2 (en) * 2002-05-21 2008-03-18 Allergan, Inc. Methods and compositions for alleviating pain
US20080091207A1 (en) * 2006-10-13 2008-04-17 Csaba Truckai Bone treatment systems and methods
US20080152709A1 (en) * 2006-12-22 2008-06-26 Drugtech Corporation Clonidine composition and method of use
US20090017090A1 (en) * 2006-07-10 2009-01-15 Arensdorf Patrick A Devices and methods for delivering active agents to the osteomeatal complex
US7507398B2 (en) * 2001-05-24 2009-03-24 Alexza Pharmaceuticals, Inc. Delivery of physiologically active compounds through an inhalation route
US7524812B2 (en) * 2003-10-02 2009-04-28 Elan Pharmaceuticals, Inc. Pharmaceutical formulation comprising ziconotide
US20090263459A1 (en) * 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc. Methods and compositions for treating intervertebral disc herniations

Family Cites Families (205)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4692147A (en) 1980-04-02 1987-09-08 Medtronic, Inc. Drug administration device
US4624255A (en) 1982-02-18 1986-11-25 Schenck Robert R Apparatus for anastomosing living vessels
US4742054A (en) 1982-11-23 1988-05-03 Naftchi Nosrat E Treatment of mammals suffering from damage to the central nervous system
US4863457A (en) 1986-11-24 1989-09-05 Lee David A Drug delivery device
US5633002A (en) 1988-10-04 1997-05-27 Boehringer Ingelheim Gmbh Implantable, biodegradable system for releasing active substance
US5077049A (en) 1989-07-24 1991-12-31 Vipont Pharmaceutical, Inc. Biodegradable system for regenerating the periodontium
US5762638A (en) 1991-02-27 1998-06-09 Shikani; Alain H. Anti-infective and anti-inflammatory releasing systems for medical devices
WO1992015286A1 (en) 1991-02-27 1992-09-17 Nova Pharmaceutical Corporation Anti-infective and anti-inflammatory releasing systems for medical devices
EP0596912A4 (en) 1991-07-16 1994-07-27 Hem Pharma Corp Modulation and diagnosis of cytokine dysfunctions.
JP2002514999A (en) 1991-09-26 2002-05-21 ヘム・ファーマスーティカル・コーポレーション Method for neutralizing IL-1 using dsRNA
US5368854A (en) 1992-08-20 1994-11-29 Schering Corporation Use of IL-10 to treat inflammatory bowel disease
US5868789A (en) 1997-02-03 1999-02-09 Huebner; Randall J. Removable suture anchor apparatus
US5522844A (en) 1993-06-22 1996-06-04 Johnson; Lanny L. Suture anchor, suture anchor installation device and method for attaching a suture to a bone
US5626838A (en) 1995-03-13 1997-05-06 The Procter & Gamble Company Use of ketorolac for treatment of squamous cell carcinomas of the oral cavity or oropharynx
WO1996033761A1 (en) 1995-04-28 1996-10-31 Medtronic, Inc. Intraparenchymal infusion catheter system
US7069634B1 (en) 1995-04-28 2006-07-04 Medtronic, Inc. Method for manufacturing a catheter
US5869079A (en) * 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
NZ311474A (en) 1995-06-09 1997-09-22 Euro Celtique Sa Formulations for providing prolonged local anesthesia
US5880343A (en) 1995-06-15 1999-03-09 Mayekawa Manufacturing Co., Ltd. Grass and method of introducing endophytic fungi into a grass
US5759583A (en) 1995-08-30 1998-06-02 Syntex (U.S.A.) Inc. Sustained release poly (lactic/glycolic) matrices
US5677321A (en) 1996-02-29 1997-10-14 Synaptic Pharmaceutical Corporation 5- and 6-(2-imidazolin-2-ylamino) and -(2-thiazolin-2-ylamino)-benzothiazoles as alpha-2 adrenergic ligands
US5711316A (en) 1996-04-30 1998-01-27 Medtronic, Inc. Method of treating movement disorders by brain infusion
US5713923A (en) 1996-05-13 1998-02-03 Medtronic, Inc. Techniques for treating epilepsy by brain stimulation and drug infusion
WO1997044057A1 (en) 1996-05-21 1997-11-27 Cytotherapeutics, Inc. System and method for delivery of cytokines using encapsulated cytokine-secreting cells
ES2339306T3 (en) 1996-11-15 2010-05-18 Cytokine Pharmasciences, Inc. USEFUL GUANILHYDRAZONES IN THE TREATMENT OF DISEASES ASSOCIATED WITH THE ACTIVATION OF CELLS T.
US20020111603A1 (en) 1996-12-02 2002-08-15 Societe De Conseils De Recherches Et D'application Device for local administration of solid or semi-solid formulations and delayed-release formulations for proposal parental administration and preparation process
FR2756493B1 (en) * 1996-12-02 2001-04-13 Delab DEVICE FOR LOCAL ADMINISTRATION OF SOLID OR SEMI-SOLID FORMULATIONS
US6495579B1 (en) 1996-12-02 2002-12-17 Angiotech Pharmaceuticals, Inc. Method for treating multiple sclerosis
WO1998024469A1 (en) 1996-12-05 1998-06-11 University Of Florida Il-10 and tgf-beta for treating autoimmune disease
US6433147B1 (en) 1997-01-28 2002-08-13 Human Genome Sciences, Inc. Death domain containing receptor-4
DE19716713A1 (en) 1997-04-21 1998-10-22 Paz Arzneimittelentwicklung Medicines containing ibuprofenthioester as inhibitors of Nf-kappaB-dependent formation of mediators of inflammation and pain
US6272370B1 (en) 1998-08-07 2001-08-07 The Regents Of University Of Minnesota MR-visible medical device for neurological interventions using nonlinear magnetic stereotaxis and a method imaging
AU7389098A (en) 1997-05-16 1998-12-08 Brigham And Women's Hospital Local anesthetic formulations
AU739384B2 (en) 1997-07-02 2001-10-11 Euro-Celtique S.A. Prolonged anesthesia in joints and body spaces
US6241736B1 (en) 1998-05-12 2001-06-05 Scimed Life Systems, Inc. Manual bone anchor placement devices
US20030077641A1 (en) 1998-03-11 2003-04-24 Laskowitz Daniel T. Methods of suppressing microglial activation and systemic inflammatory responses
US7205280B2 (en) 1998-03-11 2007-04-17 Cognosci, Inc. Methods of suppressing microglial activation
GB9806530D0 (en) 1998-03-26 1998-05-27 Glaxo Group Ltd Inflammatory mediator
US6596747B2 (en) 1998-10-29 2003-07-22 Bristol-Myers Squibb Company Compounds derived from an amine nucleus and pharmaceutical compositions comprising same
US6069008A (en) 1998-11-25 2000-05-30 Isis Pharmaceuticals Inc. Antisense modulation of NF-kappa-B p65 subunit expression
US6423321B2 (en) 1999-02-24 2002-07-23 Edward L. Tobinick Cytokine antagonists for the treatment of sensorineural hearing loss
US6419944B2 (en) 1999-02-24 2002-07-16 Edward L. Tobinick Cytokine antagonists for the treatment of localized disorders
US6177077B1 (en) 1999-02-24 2001-01-23 Edward L. Tobinick TNT inhibitors for the treatment of neurological disorders
US6982089B2 (en) 1999-02-24 2006-01-03 Tact Ip, Llc Cytokine antagonists for neurological and neuropsychiatric disorders
US20030185826A1 (en) 1999-02-24 2003-10-02 Tobinick Edward L. Cytokine antagonists for the treatment of localized disorders
US6537549B2 (en) 1999-02-24 2003-03-25 Edward L. Tobinick Cytokine antagonists for the treatment of localized disorders
US20030113318A1 (en) 1999-02-24 2003-06-19 Tobinick Edward Lewis TNF inhibition for the treatment of pre-menstrual syndrome and primary dysmenorrhea
US20030007972A1 (en) 1999-02-24 2003-01-09 Edward Tobinick Cytokine antagonists and other biologics for the treatment of bone metastases
US6436099B1 (en) 1999-04-23 2002-08-20 Sdgi Holdings, Inc. Adjustable spinal tether
WO2001008717A1 (en) 1999-08-03 2001-02-08 Smith & Nephew, Inc. Controlled release implantable devices
US6883520B2 (en) 1999-08-18 2005-04-26 Intrinsic Therapeutics, Inc. Methods and apparatus for dynamically stable spinal implant
US7951201B2 (en) 1999-10-20 2011-05-31 Anulex Technologies, Inc. Method and apparatus for the treatment of the intervertebral disc annulus
US20030144570A1 (en) 1999-11-12 2003-07-31 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating disease utilizing a combination of radioactive therapy and cell-cycle inhibitors
EP1244388B1 (en) 1999-12-06 2007-01-17 Warsaw Orthopedic, Inc. Intervertebral disc treatment device
WO2002081628A2 (en) 2001-04-05 2002-10-17 Ribozyme Pharmaceuticals, Incorporated Modulation of gene expression associated with inflammation proliferation and neurite outgrowth, using nucleic acid based technologies
US6899716B2 (en) 2000-02-16 2005-05-31 Trans1, Inc. Method and apparatus for spinal augmentation
US6287688B1 (en) 2000-03-03 2001-09-11 E. I. Du Pont De Nemours And Company Partially oriented poly(trimethylene terephthalate) yarn
US20020131954A1 (en) 2000-05-02 2002-09-19 Tobinick Edward L. Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
CA2771263A1 (en) 2000-07-27 2002-02-07 Rutgers, The State University Therapeutic polyesters and polyamides
US7087224B2 (en) 2000-10-31 2006-08-08 Amgen Inc. Method of treating anemia by administering IL-1ra
JP2004537497A (en) 2000-11-08 2004-12-16 ザ ジェネラル ホスピタル コーポレーション Methods for inhibiting pain
NZ525888A (en) 2000-12-01 2006-04-28 Max Planck Gesellschaft RNA interference mediating small RNA molecules
CA2432203C (en) 2001-01-03 2008-03-25 Michael J. Brubaker Sustained release drug delivery devices with multiple agents
US7229441B2 (en) 2001-02-28 2007-06-12 Warsaw Orthopedic, Inc. Flexible systems for spinal stabilization and fixation
US20020168360A1 (en) 2001-03-02 2002-11-14 Christine Dingivan Methods of preventing or treating inflammatory or autoimmune disorders by administering integrin alphanubeta3 antagonists in combination with other prophylactic or therapeutic agents
US8162816B2 (en) 2001-03-09 2012-04-24 Boston Scientific Scimed, Inc. System for implanting an implant and method thereof
EP1379230A4 (en) 2001-03-15 2009-01-21 Univ Pittsburgh Method of using pyruvate and/or its derivatives for the treatment of cytokine-mediated inflammatory conditions
SE0101258D0 (en) 2001-04-06 2001-04-06 A & Science Invest Ab Treatment of low back pain and whiplash associated disorder
US6632217B2 (en) 2001-04-19 2003-10-14 Microsolutions, Inc. Implantable osmotic pump
IL142933A0 (en) 2001-05-03 2002-04-21 Dentis Technology Ltd Polymeric dental implant and method for use thereof
US20050182009A1 (en) 2001-05-18 2005-08-18 Sirna Therapeutics, Inc. RNA interference mediated inhibition of NF-Kappa B / REL-A gene expression using short interfering nucleic acid (siNA)
CA2385745C (en) 2001-06-08 2015-02-17 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
US7722894B2 (en) 2001-10-22 2010-05-25 Massachusetts Institute Of Technology Biodegradable polymer
US20030157061A1 (en) 2001-12-05 2003-08-21 Pharmacia Corporation Combinations of a cyclooxygenase-2 selective inhibitor and a TNFalpha antagonist and therapeutic uses therefor
US6974462B2 (en) 2001-12-19 2005-12-13 Boston Scientific Scimed, Inc. Surgical anchor implantation device
US7399829B2 (en) 2002-01-04 2008-07-15 Xencor, Inc. Variants of RANKL protein
US8133501B2 (en) 2002-02-08 2012-03-13 Boston Scientific Scimed, Inc. Implantable or insertable medical devices for controlled drug delivery
JP2005517433A (en) 2002-02-20 2005-06-16 サーナ・セラピューティクス・インコーポレイテッド RNA interference-mediated inhibition of TNF and TNF receptor superfamily gene expression using short interfering nucleic acids (siNA)
EP1432724A4 (en) 2002-02-20 2006-02-01 Sirna Therapeutics Inc Rna interference mediated inhibition of map kinase genes
EP1487494A2 (en) 2002-02-26 2004-12-22 North Shore-Long Island Jewish Research Institute Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
AU2003225976A1 (en) 2002-03-26 2003-10-13 Centocor, Inc. Anti-tnf antibodies, compositions, methods and uses
GB2389791B (en) 2002-04-30 2006-12-13 Steven Gill Implantable drug delivery pump
AU2003237686A1 (en) 2002-05-24 2003-12-12 Max-Planck Gesellschaft Zur Forderung Der Wissenschaften E.V. Rna interference mediating small rna molecules
US7345078B2 (en) 2002-05-31 2008-03-18 The Board Of Trustees Of Michigan State University NF-κB inhibitors and uses thereof
US20040109893A1 (en) 2002-06-25 2004-06-10 Guohua Chen Sustained release dosage forms of anesthetics for pain management
US20040005569A1 (en) 2002-07-02 2004-01-08 Isis Pharmaceuticals Inc. Antisense modulation of NF-kappa-B p50 subunit expression
US7283956B2 (en) 2002-09-18 2007-10-16 Motorola, Inc. Noise suppression
US7166641B2 (en) 2002-10-02 2007-01-23 Yung Shin Pharmaceutical Industrial Co., Ltd. Pharmaceutically acceptable salts containing local anesthetic and anti-inflammatory activities and methods for preparing the same
EP1556011A1 (en) 2002-10-28 2005-07-27 Polymerix Corporation Therapeutic compositions
EP2213738B1 (en) 2002-11-14 2012-10-10 Dharmacon, Inc. siRNA molecules targeting Bcl-2
US6877763B2 (en) 2002-12-24 2005-04-12 Phillip J. Ulrich Infant sports stroller
US20050048099A1 (en) 2003-01-09 2005-03-03 Allergan, Inc. Ocular implant made by a double extrusion process
EP1462111A1 (en) 2003-03-28 2004-09-29 Universiteit Utrecht Holding B.V. Composition for inducing immunotolerance
EP1613266A4 (en) 2003-04-15 2009-05-06 Theraquest Biosciences Llc Methods of treating pain and compositions for use therefor
US7288084B2 (en) 2003-04-28 2007-10-30 Boston Scientific Scimed, Inc. Drug-loaded medical device
US20040224893A1 (en) 2003-05-06 2004-11-11 Li-Hsien Wang Methods of using IL-1 antagonists to treat neointimal hyperplasia
WO2004108111A1 (en) 2003-05-30 2004-12-16 Alza Corporation Implantable elastomeric depot compositions, uses thereof and method of manufacturing
US7141597B2 (en) 2003-09-12 2006-11-28 Allergan, Inc. Nonsedating α-2 agonists
US7144412B2 (en) 2003-06-25 2006-12-05 Wolf Medical Enterprises, Inc. Gold suture and method of use in wound closure
ES2291893T3 (en) 2003-06-26 2008-03-01 Mediolanum Pharmaceuticals Limited SUBCUTANEOUS IMPLANTS WITH LIMITED INITIAL RELEASE OF THE ACTIVE AND SUBSEQUENT PRINCIPLE LINEARLY VARIABLE LONG RELEASE OF THE SAME.
US20050026979A1 (en) 2003-07-31 2005-02-03 Maha Ghazzi Methods for treating inflammation and inflammation-associated diseases with a statin and ether
WO2005034998A2 (en) 2003-09-12 2005-04-21 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
US7418292B2 (en) 2003-10-01 2008-08-26 Medtronic, Inc. Device and method for attenuating an immune response
US20050187176A1 (en) 2003-10-10 2005-08-25 Bates Paula J. Method for inhibiting NF-kappa B signaling and use to treat or prevent human diseases
US20050245557A1 (en) 2003-10-15 2005-11-03 Pain Therapeutics, Inc. Methods and materials useful for the treatment of arthritic conditions, inflammation associated with a chronic condition or chronic pain
US20050090549A1 (en) 2003-10-23 2005-04-28 Medtronic, Inc. Intrathecal gabapentin for treatment of pain
CA2543779A1 (en) 2003-10-24 2005-05-06 Medtronic, Inc. Techniques to treat neurological disorders by attenuating the production of pro-inflammatory mediators
US20050129731A1 (en) 2003-11-03 2005-06-16 Roland Horres Biocompatible, biostable coating of medical surfaces
CA2545062A1 (en) 2003-11-06 2005-05-26 Research Development Foundation Selective inhibitors of nuclear factor-kb activation and uses thereof
US20050175709A1 (en) 2003-12-11 2005-08-11 Baty Ace M.Iii Therapeutic microparticles
SE0303397D0 (en) 2003-12-17 2003-12-17 Index Pharmaceuticals Ab Compounds and method for RNA interference
AU2005209331A1 (en) 2004-01-30 2005-08-11 Peplin Biolipids Pty Ltd Therapeutic and carrier molecules
US7232435B2 (en) 2004-02-06 2007-06-19 Medtronic, Inc. Delivery of a sympatholytic cardiovascular agent to the central nervous system to counter heart failure and pathologies associated with heart failure
US20050197312A1 (en) 2004-03-03 2005-09-08 Kevin Fitzgerald Transcription factor RNA interference reagents and methods of use thereof
US7717848B2 (en) 2004-03-16 2010-05-18 Medtronic, Inc. Collecting sleep quality information via a medical device
US7361168B2 (en) 2004-04-21 2008-04-22 Acclarent, Inc. Implantable device and methods for delivering drugs and other substances to treat sinusitis and other disorders
US7410480B2 (en) 2004-04-21 2008-08-12 Acclarent, Inc. Devices and methods for delivering therapeutic substances for the treatment of sinusitis and other disorders
US8147865B2 (en) 2004-04-30 2012-04-03 Allergan, Inc. Steroid-containing sustained release intraocular implants and related methods
US8685435B2 (en) * 2004-04-30 2014-04-01 Allergan, Inc. Extended release biodegradable ocular implants
US7589057B2 (en) 2004-04-30 2009-09-15 Allergan, Inc. Oil-in-water method for making alpha-2 agonist polymeric drug delivery systems
WO2006022611A2 (en) 2004-06-26 2006-03-02 Binnur Ozturk Neuropathy cream
US8512730B2 (en) 2004-07-12 2013-08-20 Isto Technologies, Inc. Methods of tissue repair and compositions therefor
US20060046960A1 (en) 2004-09-02 2006-03-02 Mckay William F Controlled and directed local delivery of anti-inflammatory compositions
US20060046961A1 (en) 2004-09-02 2006-03-02 Mckay William F Controlled and directed local delivery of anti-inflammatory compositions
US20060074422A1 (en) 2004-09-27 2006-04-06 Story Brooks J Suture anchor and void filler combination
US20060253100A1 (en) 2004-10-22 2006-11-09 Medtronic, Inc. Systems and Methods to Treat Pain Locally
US20060183786A1 (en) 2005-02-16 2006-08-17 Chi Mei Foundation Hospital Injectable long-acting analgesic composition comprising an ester derivative of ketorolac
US20060204548A1 (en) 2005-03-01 2006-09-14 Allergan, Inc. Microimplants for ocular administration
US20060211665A1 (en) 2005-03-17 2006-09-21 Ranawat Chitranjan S Reduction of postoperative pain medication
WO2006104913A2 (en) 2005-03-25 2006-10-05 Medtronic, Inc. USE OF ANTI-TNF OR ANTI-ILl RNAI TO SUPPRESS PRO- INFLAMMATORY CYTOKINE ACTIONS LOCALLY TO TREAT PAIN
US7767656B2 (en) 2005-04-25 2010-08-03 Molly S Shoichet Blends of temperature sensitive and anionic polymers for drug delivery
AU2006249684A1 (en) 2005-05-20 2006-11-30 Omeros Corporation Cyclooxygenase inhibitor and calcium channel antagonist compositions and methods for use in urological procedures
US20070000509A1 (en) * 2005-06-14 2007-01-04 Mays Carol A Hair volumizing ornament and method
US20090182026A1 (en) 2005-06-29 2009-07-16 Allergan, Inc. Alpha 2 adrenergic agonists
US8051414B2 (en) * 2005-08-01 2011-11-01 Siemens Aktiengesellschaft Method and system for remote software updating of a medical device
WO2007098443A2 (en) 2006-02-17 2007-08-30 Waseet Vance Apparatuses and techniques for bioactive drug delivery in the prostate gland
US7964585B2 (en) 2006-03-14 2011-06-21 Case Western Reserve University Composition and method of treating peripheral neuropathy
US20070253994A1 (en) 2006-04-28 2007-11-01 Medtronic, Inc. Intraspinal Drug Delivery Methods and Devices To Alleviate Chronic Pelvic Pain
US20080021074A1 (en) 2006-06-29 2008-01-24 Questcor Pharmaceuticals, Inc. Pharmaceutical Compositions and Related Methods of Treatment
US7628408B2 (en) 2006-07-14 2009-12-08 Timothy Kolesa System and method for moving material past an obstacle
WO2008009141A1 (en) 2006-07-21 2008-01-24 Queen's University At Kingston Methods and therapies for potentiating a therapeutic action of an alpha- 2 adrenergic receptor agonist and inhibiting and/or reversing tolerance to alpha- 2 adrenergic receptor agonists
US20080075777A1 (en) 2006-07-31 2008-03-27 Kennedy Michael T Apparatus and methods for preparing solid particles
US20080095831A1 (en) 2006-08-10 2008-04-24 Mc Graw Thomas L Topical formulation of multilamellar vesicles composition for percutaneous absorption of pharmaceutically active agent
US20080317805A1 (en) 2007-06-19 2008-12-25 Mckay William F Locally administrated low doses of corticosteroids
ITBO20070461A1 (en) 2007-07-04 2009-01-05 Dinamica Generale S R L SYSTEM TO CHECK THE LOADING OF ONE OR MORE FOODS IN A MIXED MIXING UNIT BY MEANS OF A MECHANICAL SHOVEL MOUNTED ON A MOTOR VEHICLE
US8470360B2 (en) 2008-04-18 2013-06-25 Warsaw Orthopedic, Inc. Drug depots having different release profiles for reducing, preventing or treating pain and inflammation
US8029478B2 (en) 2007-10-31 2011-10-04 Warsaw Orthopedic, Inc. Implantable device and method for delivering drug depots to a site beneath the skin
WO2009100441A2 (en) 2008-02-08 2009-08-13 Impax Laboratories, Inc. Depot formulations
US20090264489A1 (en) 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc. Method for Treating Acute Pain with a Formulated Drug Depot in Combination with a Liquid Formulation
US9289409B2 (en) 2008-04-18 2016-03-22 Warsaw Orthopedic, Inc. Sulindac formulations in a biodegradable material
US20090264478A1 (en) 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc. Sulfasalazine formulations in a biodegradable polymer carrier
US9072727B2 (en) 2008-04-18 2015-07-07 Warsaw Orthopedic, Inc. Alpha adrenergic receptor agonists for treatment of degenerative disc disease
US8722079B2 (en) 2008-04-18 2014-05-13 Warsaw Orthopedic, Inc. Methods for treating conditions such as dystonia and post-stroke spasticity with clonidine
US8846068B2 (en) 2008-04-18 2014-09-30 Warsaw Orthopedic, Inc. Methods and compositions for treating post-operative pain comprising a local anesthetic
US8889173B2 (en) 2008-04-18 2014-11-18 Warsaw Orthopedic, Inc. Alpha adrenergic receptor agonists for treatment of pain and/or inflammation
US20090264477A1 (en) 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc., An Indiana Corporation Beta adrenergic receptor agonists for treatment of pain and/or inflammation
US8946277B2 (en) 2008-04-18 2015-02-03 Warsaw Orthopedic, Inc. Clonidine formulations in a biodegradable polymer carrier
US8557273B2 (en) 2008-04-18 2013-10-15 Medtronic, Inc. Medical devices and methods including polymers having biologically active agents therein
US8629172B2 (en) 2008-04-18 2014-01-14 Warsaw Orthopedic, Inc. Methods and compositions for treating post-operative pain comprising clonidine
US20090263489A1 (en) 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc. Analgesic and anti-inflammatory compositions and methods for reducing, preventing or treating pain and inflammation
US9610243B2 (en) 2008-04-18 2017-04-04 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable polymer
US8956641B2 (en) 2008-04-18 2015-02-17 Warsaw Orthopedic, Inc. Alpha adrenergic receptor agonists for treatment of inflammatory diseases
US8420114B2 (en) 2008-04-18 2013-04-16 Warsaw Orthopedic, Inc. Alpha and beta adrenergic receptor agonists for treatment of pain and / or inflammation
US8956642B2 (en) 2008-04-18 2015-02-17 Medtronic, Inc. Bupivacaine formulation in a polyorthoester carrier
US9132085B2 (en) 2008-04-18 2015-09-15 Warsaw Orthopedic, Inc. Compositions and methods for treating post-operative pain using clonidine and bupivacaine
US20090263443A1 (en) 2008-04-18 2009-10-22 Warsaw Orthopedics, Inc. Methods for treating post-operative effects such as spasticity and shivering with clondine
US7993666B2 (en) 2008-04-18 2011-08-09 Warsaw Orthopedic, Inc. Methods and compositions for treating pain comprising a statin
US8940315B2 (en) 2008-04-18 2015-01-27 Medtronic, Inc. Benzodiazepine formulation in a polyorthoester carrier
US8524267B2 (en) 2008-04-18 2013-09-03 Warsaw Orthopedic, Inc. Dexamethasone formulations in a biodegradable material
US9125917B2 (en) 2008-04-18 2015-09-08 Warsaw Orthopedic, Inc. Fluocinolone formulations in a biodegradable polymer carrier
US8956636B2 (en) 2008-04-18 2015-02-17 Warsaw Orthopedic, Inc. Methods and compositions for treating postoperative pain comprosing ketorolac
US20090263451A1 (en) 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc. Anti-Inflammatory and/or Analgesic Agents for Treatment of Myofascial Pain
US9132119B2 (en) 2008-04-18 2015-09-15 Medtronic, Inc. Clonidine formulation in a polyorthoester carrier
US20100015049A1 (en) 2008-07-16 2010-01-21 Warsaw Orthopedic, Inc. Methods and compositions for treating postoperative pain comprising nonsteroidal anti-inflammatory agents
US8557271B2 (en) 2008-07-16 2013-10-15 Warsaw Orthopedic, Inc. Drug depot implantable within a joint
US8202531B2 (en) * 2008-07-23 2012-06-19 Warsaw Orthopedic, Inc. Drug depots having one or more anchoring members
US20100098746A1 (en) 2008-10-20 2010-04-22 Warsaw Orthopedic, Inc. Compositions and methods for treating periodontal disease comprising clonidine, sulindac and/or fluocinolone
US9623222B2 (en) 2008-10-30 2017-04-18 Warsaw Orthopedic, Inc. Drug depot with anchor
US8822546B2 (en) 2008-12-01 2014-09-02 Medtronic, Inc. Flowable pharmaceutical depot
US8980317B2 (en) 2008-12-23 2015-03-17 Warsaw Orthopedic, Inc. Methods and compositions for treating infections comprising a local anesthetic
US20100203102A1 (en) 2009-02-10 2010-08-12 Warsaw Orthopedic, Inc. Compositions and methods for treating post-operative pain using bupivacaine and an anti-onflammatory agent
US20100228097A1 (en) 2009-03-04 2010-09-09 Warsaw Orthopedic, Inc. Methods and compositions to diagnose pain
US20100239632A1 (en) 2009-03-23 2010-09-23 Warsaw Orthopedic, Inc. Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
US8617583B2 (en) 2009-07-17 2013-12-31 Warsaw Orthopedic, Inc. Alpha adrenergic receptor agonists for prevention or treatment of a hematoma, edema, and/or deep vein thrombosis
US20110027331A1 (en) 2009-07-29 2011-02-03 Warsaw Orthopedic, Inc. An implantable drug depot having a reversible phase transition material for treatment of pain and/or inflammation
US8231891B2 (en) 2009-07-31 2012-07-31 Warsaw Orthopedic, Inc. Implantable drug depot for weight control
WO2011044229A1 (en) 2009-10-06 2011-04-14 Allergan, Inc. 2h-pyrrol-5-amine derivatives as alpha adrenergic receptor modulators
US20110097375A1 (en) 2009-10-26 2011-04-28 Warsaw Orthopedic, Inc. Formulation for preventing or reducing bleeding at a surgical site
US20110097380A1 (en) 2009-10-28 2011-04-28 Warsaw Orthopedic, Inc. Clonidine formulations having antimicrobial properties
US20110106110A1 (en) 2009-10-30 2011-05-05 Warsaw Orthopedic, Inc. Devices and methods for implanting a plurality of drug depots having one or more anchoring members
US9050274B2 (en) 2010-01-28 2015-06-09 Warsaw Orthopedic, Inc. Compositions and methods for treating an intervertebral disc using bulking agents or sealing agents
WO2011133370A1 (en) 2010-04-23 2011-10-27 Medtronic, Inc. Shelf stable pharmaceutical depot
WO2011139595A2 (en) 2010-04-27 2011-11-10 Medtronic, Inc. Elongated biodegradable depot for sustained drug release to treat chronic pelvic pain
US20120029042A1 (en) 2010-07-30 2012-02-02 Warsaw Orthopedic, Inc. Clonidine for treatment of bone cancer
WO2012075451A2 (en) 2010-12-03 2012-06-07 Warsaw Orthopedic, Inc. Clonidine and gaba compounds in a biodegradable polymer carrier
US20120142747A1 (en) 2010-12-03 2012-06-07 Warsaw Orthopedic, Inc. Compositions and methods for delivering clonidine to a target tissue site
US20120142648A1 (en) 2010-12-03 2012-06-07 Warsaw Orthopedic, Inc. Methods for delivering clonidine compositions in biodegradable polymer carrier and local steriods to a target tissue site
WO2012075447A2 (en) 2010-12-03 2012-06-07 Warsaw Orthopedic, Inc. Compositions and methods for delivering clonidine and bupivacaine to a target tissue site
US9005634B2 (en) 2011-04-13 2015-04-14 Medtronic, Inc. Shelf stable pharmaceutical depot
US9211274B2 (en) 2012-02-01 2015-12-15 Warsaw Orthopedic, Inc. TRPV1 compounds in a biodegradable polymer carrier
US20130217673A1 (en) 2012-02-22 2013-08-22 Warsaw Orthopedic, Inc Mixed monoamine reuptake inhibitor in a biodegradable polymer carrier
US9511018B2 (en) 2012-04-05 2016-12-06 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable matrix
US8735504B2 (en) 2012-05-02 2014-05-27 Warsaw Orthopedic, Inc. Methods for preparing polymers having low residual monomer content
US9066853B2 (en) 2013-01-15 2015-06-30 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable fiber
US20150342964A1 (en) 2014-05-30 2015-12-03 Warsaw Orthopedic, Inc. Dexamethasone polymer implant for pain

Patent Citations (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3020660A (en) * 1959-11-30 1962-02-13 Scherotto John Collapsible imitation tree
US3190802A (en) * 1961-10-09 1965-06-22 Boehringer Sohn Ingelheim Shaving composition and method of using same
US4765974A (en) * 1985-12-24 1988-08-23 Nitto Electric Industrial Co., Ltd. Preparation for percutaneous administration
US5175052A (en) * 1988-05-11 1992-12-29 Nitto Denko Corporation Adhesive tape preparation of clonidine
US5447947A (en) * 1990-02-26 1995-09-05 Arc 1 Compositions and methods of treatment of sympathetically maintained pain
US5484607A (en) * 1993-10-13 1996-01-16 Horacek; H. Joseph Extended release clonidine formulation
US5869100A (en) * 1993-10-13 1999-02-09 Horacek; H. Joseph Extended release clonidine formulation (tablet)
US6030642A (en) * 1993-10-13 2000-02-29 Horacek; H. Joseph Extended release clonidine formulation (capsule)
US5635204A (en) * 1994-03-04 1997-06-03 Montefiore Medical Center Method for transdermal induction of anesthesia, analgesia or sedation
US5980927A (en) * 1995-02-10 1999-11-09 Medtronic, Inc. Method and apparatus for administering analgesics, and method for making same device
US5942503A (en) * 1995-11-14 1999-08-24 Boehringer Indelheim Kg Use of Epinastine for the treatment of pain
US5900245A (en) * 1996-03-22 1999-05-04 Focal, Inc. Compliant tissue sealants
US5945416A (en) * 1996-03-25 1999-08-31 Eli Lilly And Company Method for treating pain
US6331311B1 (en) * 1996-12-20 2001-12-18 Alza Corporation Injectable depot gel composition and method of preparing the composition
US5801188A (en) * 1997-01-08 1998-09-01 Medtronic Inc. Clonidine therapy enhancement
US20020009454A1 (en) * 1997-02-10 2002-01-24 Amgen Inc. Composition and method for treating inflammatory diseases
US20010018075A1 (en) * 1997-07-15 2001-08-30 Takada Shigeyuki Process for producing sustained-release preparation
US5942530A (en) * 1997-08-28 1999-08-24 Eli Lilly And Company Method for treating pain
US6428804B1 (en) * 1997-10-27 2002-08-06 Ssp Co., Ltd. Intra-articular preparation for the treatment of arthropathy
US6069129A (en) * 1998-03-13 2000-05-30 Mrs, Llc Elastin derived composition and method of using same
US20020064547A1 (en) * 1998-03-19 2002-05-30 Rey T. Chern Liquid polymeric compositions for controlled release of bioactive substances
US6451335B1 (en) * 1998-07-02 2002-09-17 Euro-Celtique S.A. Formulations and methods for providing prolonged local anesthesia
US6277413B1 (en) * 1998-07-17 2001-08-21 Skyepharma, Inc. Biodegradable compositions for the controlled release of encapsulated substances
US6632457B1 (en) * 1998-08-14 2003-10-14 Incept Llc Composite hydrogel drug delivery systems
US6179862B1 (en) * 1998-08-14 2001-01-30 Incept Llc Methods and apparatus for in situ formation of hydrogels
US6630155B1 (en) * 1998-10-28 2003-10-07 Atrix Laboratories, Inc. Controlled release liquid delivery compositions with low initial drug burst
US6773714B2 (en) * 1998-10-28 2004-08-10 Atrix Laboratories, Inc. Polymeric delivery formulations of leuprolide with improved efficacy
US6287588B1 (en) * 1999-04-29 2001-09-11 Macromed, Inc. Agent delivering system comprised of microparticle and biodegradable gel with an improved releasing profile and methods of use thereof
US6306424B1 (en) * 1999-06-30 2001-10-23 Ethicon, Inc. Foam composite for the repair or regeneration of tissue
US6534048B1 (en) * 1999-10-26 2003-03-18 Curatek Pharmaceuticals Holding, Inc. Topical clonidine preparation
US6147102A (en) * 1999-10-26 2000-11-14 Curatek Pharmaceuticals Holding, Inc. Clonidine preparations
US6461631B1 (en) * 1999-11-16 2002-10-08 Atrix Laboratories, Inc. Biodegradable polymer composition
US20020090398A1 (en) * 1999-11-16 2002-07-11 Atrix Laboratories, Inc. Biodegradable polymer composition
US20040028726A1 (en) * 1999-11-29 2004-02-12 Wilfried Fischer Transdermal systems for the delivery of clonidine
US6589549B2 (en) * 2000-04-27 2003-07-08 Macromed, Incorporated Bioactive agent delivering system comprised of microparticles within a biodegradable to improve release profiles
US20020058656A1 (en) * 2000-09-19 2002-05-16 Ockert David M. Triple drug therapy for the treatment and prevention of acute or chronic pain
US6417184B1 (en) * 2000-09-19 2002-07-09 David M. Ockert Triple drug therapy for the treatment and prevention of acute or chronic pain
US20020094998A1 (en) * 2000-11-01 2002-07-18 Burke James A. Methods and compositions for treatment of ocular neovascularization and neural injury
US20030009145A1 (en) * 2001-03-23 2003-01-09 Struijker-Boudier Harry A.J. Delivery of drugs from sustained release devices implanted in myocardial tissue or in the pericardial space
US20030022926A1 (en) * 2001-05-07 2003-01-30 Lavand'homme Patricia Method for treating neuropathic pain and pharmaceutical preparation therefor
US7507398B2 (en) * 2001-05-24 2009-03-24 Alexza Pharmaceuticals, Inc. Delivery of physiologically active compounds through an inhalation route
US6992110B2 (en) * 2001-11-05 2006-01-31 Cypress Bioscience, Inc. Methods of treating fibromyalgia syndrome, chronic fatigue syndrome and pain
US20040082540A1 (en) * 2001-11-13 2004-04-29 Hermida Ochoa Elias Humberto Use of a mixture of sodium hyaluronate and chondroitin sulfate for the treatment of osteoarthritis
US20040214793A1 (en) * 2001-11-13 2004-10-28 Hermida Ochoa Elias Humberto Regeneration of articular cartilage damaged by grade i and ii osteoarthritis by means of the intraarticular application of a mixture of sodium hyaluronate and chondroitin sulfate in a gel vehicle
US20030185872A1 (en) * 2002-03-27 2003-10-02 Frank Kochinke Methods and drug delivery systems for the treatment of orofacial diseases
US7345065B2 (en) * 2002-05-21 2008-03-18 Allergan, Inc. Methods and compositions for alleviating pain
US20040072799A1 (en) * 2002-07-19 2004-04-15 Omeros Corporation Biodegradable triblock copolymers, synthesis methods therefore, and hydrogels and biomaterials made there from
US20050197293A1 (en) * 2002-10-28 2005-09-08 Scott Mellis Use of an IL-1 antagonist for treating arthritis
US20040101582A1 (en) * 2002-11-25 2004-05-27 Richard Wolicki Treatment of neuropathy
US20070202074A1 (en) * 2003-01-15 2007-08-30 Shalaby Shalaby W Polymeric precursors of non-absorbable, in situ-forming hydrogels and applications thereof
US20050043706A1 (en) * 2003-03-14 2005-02-24 Eaton Donald J. Sinus delivery of sustained release therapeutics
US20040208917A1 (en) * 2003-04-16 2004-10-21 Wilfried Fischer Transdermal systems for the release of clonidine
US20050095277A1 (en) * 2003-06-25 2005-05-05 Binnur Ozturk Neuropathy cream
US20040265364A1 (en) * 2003-06-25 2004-12-30 Binnur Ozturk Neuropathy cream
US20080171075A1 (en) * 2003-06-25 2008-07-17 Binnur Ozturk Neuropathy cream
US20050058696A1 (en) * 2003-09-12 2005-03-17 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
US20050059744A1 (en) * 2003-09-12 2005-03-17 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
US7524812B2 (en) * 2003-10-02 2009-04-28 Elan Pharmaceuticals, Inc. Pharmaceutical formulation comprising ziconotide
US20050142163A1 (en) * 2003-11-10 2005-06-30 Angiotech International Ag Medical implants and fibrosis-inducing agents
US20050186261A1 (en) * 2004-01-30 2005-08-25 Angiotech International Ag Compositions and methods for treating contracture
US20050245905A1 (en) * 2004-04-30 2005-11-03 Schmidt Steven P Local drug-delivery system
US20050288620A1 (en) * 2004-06-28 2005-12-29 Shippert Ronald D Nose pack method and apparatus
US20060148903A1 (en) * 2004-11-24 2006-07-06 Algorx Pharmaceuticals, Inc. Capsaicinoid gel formulation and uses thereof
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060189944A1 (en) * 2005-02-08 2006-08-24 Campbell Patrick K Spray for fluent materials
US20070005094A1 (en) * 2005-04-04 2007-01-04 Eaton Donald J Device and methods for treating paranasal sinus conditions
US20070021358A1 (en) * 2005-05-25 2007-01-25 Elazer Edelman Localized delivery of cardiac inotropic agents
US20070066864A1 (en) * 2005-09-06 2007-03-22 Sean Forde Devices and methods for treating cardiac tissue
US20070104769A1 (en) * 2005-11-04 2007-05-10 Lifescience Plus, Inc. Bioabsorbable hemostatic gauze
US20070156180A1 (en) * 2005-12-30 2007-07-05 Jaax Kristen N Methods and systems for treating osteoarthritis
US20070178138A1 (en) * 2006-02-01 2007-08-02 Allergan, Inc. Biodegradable non-opthalmic implants and related methods
US20070243225A1 (en) * 2006-04-13 2007-10-18 Mckay William F Drug depot implant designs and methods of implantation
US20070243228A1 (en) * 2006-04-13 2007-10-18 Mckay William F Drug depot implant designs and methods of implantation
US20080009830A1 (en) * 2006-06-27 2008-01-10 Fujimoto Kazuro L Biodegradable elastomeric patch for treating cardiac or cardiovascular conditions
US20090017090A1 (en) * 2006-07-10 2009-01-15 Arensdorf Patrick A Devices and methods for delivering active agents to the osteomeatal complex
US20080038351A1 (en) * 2006-08-14 2008-02-14 Neal Beals Flowable carrier matrix
US20080091207A1 (en) * 2006-10-13 2008-04-17 Csaba Truckai Bone treatment systems and methods
US20080152709A1 (en) * 2006-12-22 2008-06-26 Drugtech Corporation Clonidine composition and method of use
US20090263459A1 (en) * 2008-04-18 2009-10-22 Warsaw Orthopedic, Inc. Methods and compositions for treating intervertebral disc herniations

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8449607B2 (en) 2007-12-18 2013-05-28 Cormatrix Cardiovascular, Inc. Prosthetic tissue valve
US8679176B2 (en) 2007-12-18 2014-03-25 Cormatrix Cardiovascular, Inc Prosthetic tissue valve
USRE48948E1 (en) 2008-04-18 2022-03-01 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable polymer
US10653619B2 (en) 2009-03-23 2020-05-19 Medtronic, Inc. Drug depots for treatment of pain and inflammation
US20140221447A1 (en) * 2009-10-26 2014-08-07 Warsaw Orthopedic, Inc. Formulation for preventing or reducing bleeding at a surgical site
US9358223B2 (en) * 2009-10-26 2016-06-07 Warsaw Orthopedic, Inc. Formulation for preventing or reducing bleeding at a surgical site
US9861590B2 (en) * 2010-10-19 2018-01-09 Covidien Lp Self-supporting films for delivery of therapeutic agents
JP2012087121A (en) * 2010-10-19 2012-05-10 Tyco Healthcare Group Lp Method of forming self-supporting film for delivery of therapeutic agent
JP2012087122A (en) * 2010-10-19 2012-05-10 Tyco Healthcare Group Lp Self-supporting film for delivery of therapeutic agent
US20120095418A1 (en) * 2010-10-19 2012-04-19 Joshua Stopek Self-Supporting Films For Delivery Of Therapeutic Agents
JP2014500320A (en) * 2010-12-20 2014-01-09 コーマトリックス カーディオバスキュラー, インコーポレイテッド Patch extractant for localized tissue disease or defect area of the present invention
WO2012087606A1 (en) * 2010-12-20 2012-06-28 Saint Joseph's Translational Research Institute, Inc. A drug eluting patch for the treatment of localized tissue disease or defect
AU2011349853B2 (en) * 2010-12-20 2017-02-23 Cormatrix Cardiovascular, Inc. A drug eluting patch for the treatment of localized tissue disease or defect
US9511077B2 (en) 2011-04-25 2016-12-06 Warsaw Orthopedic, Inc. Medical devices and methods comprising an anabolic agent for wound healing
US9592243B2 (en) 2011-04-25 2017-03-14 Warsaw Orthopedic, Inc. Medical devices and methods comprising an anabolic agent for treatment of an injury
WO2012148570A1 (en) * 2011-04-25 2012-11-01 Warsaw Orthopedic, Inc. Medical devices and methods comprising an anabolic agent for wound healing
US8845719B2 (en) 2011-05-27 2014-09-30 Cormatrix Cardiovascular, Inc Extracellular matrix material conduits and methods of making and using same
US8696744B2 (en) 2011-05-27 2014-04-15 Cormatrix Cardiovascular, Inc. Extracellular matrix material valve conduit and methods of making thereof
WO2012166539A1 (en) * 2011-05-31 2012-12-06 Cormatrix Cardiovascular, Inc. Compositions for preventing cardiac arrhythmia
US9566241B2 (en) * 2012-02-21 2017-02-14 Auburn University Buprenorphine nanoparticle composition and methods thereof
US10154968B2 (en) * 2012-02-21 2018-12-18 Auburn University Buprenorphine nanoparticle composition and methods thereof
US20160008286A1 (en) * 2012-02-21 2016-01-14 Auburn University Buprenorphine nanoparticle composition and methods thereof
US20160354992A1 (en) * 2015-06-02 2016-12-08 Biotronik Se & Co. Kg Therapeutic Or Diagnostic Medical Product Having An Adhesion-Enhancing Surface Structure
US11654250B2 (en) 2015-08-26 2023-05-23 Medtronic Xomed, Inc. Resorbable, drug-eluting submucosal turbinate implant device and method
US10596330B2 (en) 2015-08-26 2020-03-24 Medtronic Xomed, Inc. Resorbable, drug-eluting submucosal turbinate implant device and method
JP2019500944A (en) * 2015-12-08 2019-01-17 メドトロニック・ゾーメド・インコーポレーテッド Dissolvable nasal sinus sponge
US11045577B2 (en) 2015-12-08 2021-06-29 Medtronic Xomed, Inc. Dissolvable nasal sinus sponge
US11839693B2 (en) 2015-12-08 2023-12-12 Medtronic Xomed, Inc. Dissolvable nasal sinus sponge
US11013750B2 (en) * 2017-01-20 2021-05-25 Aurin Biotech Inc. Methods for treating and preventing nociceptive pain
US20180207180A1 (en) * 2017-01-20 2018-07-26 Aurin Biotech Inc. Methods for treating and preventing nociceptive pain
WO2019060549A1 (en) * 2017-09-20 2019-03-28 Sinus Clear Innovations, Llc Wound healing device and methods for wound healing

Also Published As

Publication number Publication date
WO2010111178A4 (en) 2011-03-24
EP2410998A2 (en) 2012-02-01
WO2010111178A3 (en) 2011-02-03
US20190110982A1 (en) 2019-04-18
US10653619B2 (en) 2020-05-19
EP2410998A4 (en) 2012-08-15
US20170014337A1 (en) 2017-01-19
WO2010111178A2 (en) 2010-09-30

Similar Documents

Publication Publication Date Title
US10653619B2 (en) Drug depots for treatment of pain and inflammation
US9265733B2 (en) Drug depots having different release profiles for reducing, preventing or treating pain and inflammation
US9351959B2 (en) Alpha adreneric receptor agonists for treatment of degenerative disc disease
US20110027331A1 (en) An implantable drug depot having a reversible phase transition material for treatment of pain and/or inflammation
US20090263489A1 (en) Analgesic and anti-inflammatory compositions and methods for reducing, preventing or treating pain and inflammation
US20110106110A1 (en) Devices and methods for implanting a plurality of drug depots having one or more anchoring members
US9623222B2 (en) Drug depot with anchor
US8420114B2 (en) Alpha and beta adrenergic receptor agonists for treatment of pain and / or inflammation
AU2009236152B2 (en) Alpha adrenergic receptor agonists for treatment of pain and/or inflammation
US8617583B2 (en) Alpha adrenergic receptor agonists for prevention or treatment of a hematoma, edema, and/or deep vein thrombosis
US20090264477A1 (en) Beta adrenergic receptor agonists for treatment of pain and/or inflammation

Legal Events

Date Code Title Description
AS Assignment

Owner name: MEDTRONIC, INC., MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WALSH, ANDREW J. LOWENTHAL;REEL/FRAME:022474/0453

Effective date: 20090311

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: COMPANION SPINE, LLC, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MEDTRONIC INC.;REEL/FRAME:054787/0479

Effective date: 20200925

AS Assignment

Owner name: COMPANION SPINE, LLC, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MEDTRONIC INC.;REEL/FRAME:057502/0123

Effective date: 20200925