WO2016010771A1 - Immediate release abuse deterrent liquid fill dosage form - Google Patents

Immediate release abuse deterrent liquid fill dosage form Download PDF

Info

Publication number
WO2016010771A1
WO2016010771A1 PCT/US2015/039336 US2015039336W WO2016010771A1 WO 2016010771 A1 WO2016010771 A1 WO 2016010771A1 US 2015039336 W US2015039336 W US 2015039336W WO 2016010771 A1 WO2016010771 A1 WO 2016010771A1
Authority
WO
WIPO (PCT)
Prior art keywords
peg
capsule
active substance
daltons
cst
Prior art date
Application number
PCT/US2015/039336
Other languages
French (fr)
Inventor
Edwin R. THOMPSON
Eric R. THOMPSON
Nicholas R. MYSLINKI
Steven F. KEMENY
Matthew N. HART
Original Assignee
Pharmaceutical Manufacturing Research Services, 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 Pharmaceutical Manufacturing Research Services, Inc. filed Critical Pharmaceutical Manufacturing Research Services, Inc.
Priority to ES15821947T priority Critical patent/ES2809458T3/en
Priority to CA2955229A priority patent/CA2955229C/en
Priority to AU2015290098A priority patent/AU2015290098B2/en
Priority to JP2017502652A priority patent/JP6371463B2/en
Priority to EP15821947.7A priority patent/EP3169315B1/en
Priority to DK15821947.7T priority patent/DK3169315T3/en
Publication of WO2016010771A1 publication Critical patent/WO2016010771A1/en
Priority to AU2019200026A priority patent/AU2019200026A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4833Encapsulating processes; Filling of capsules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids

Definitions

  • the present disclosure relates to an oral immediate release, abuse deterrent dosage form.
  • the dosage form contains polyethylene glycol (PEG) to reduce abuse by non-oral administration routes, e.g. intranasal and/or intravenous.
  • PEG polyethylene glycol
  • the composition of PEG is designed to allow for immediate release of the active ingredient while deterring abuse and maintaining stability of the dosage form at elevated temperatures.
  • FDA-approved drugs are provided in many different forms based on the type of active substance, the indication treated and the preferred route of administration. These forms include enteral formulations (e.g., tablets, capsules or pills), parenteral formulations (e.g., injectable formulations such as intravenous, subcutaneous, intramuscular and intraarticular), liquid formulations (e.g., elixirs), lyophilized formulations and topical formulations.
  • enteral formulations e.g., tablets, capsules or pills
  • parenteral formulations e.g., injectable formulations such as intravenous, subcutaneous, intramuscular and intraarticular
  • liquid formulations e.g., elixirs
  • lyophilized formulations e.g., lyophilized formulations and topical formulations.
  • a majority of the FDA-approved drugs are currently available in enteral form, as either a tablet or capsule.
  • U.S. 2014/0010873 (assigned to Egalet Ltd.) is directed to an abuse-deterrent pharmaceutical composition including at least one polyethylene oxide and at least one plasticizer.
  • the polyethylene oxide has an average molecular weight of at least 1 ,000,000 Daltons, and the pharmaceutical composition includes at least 5 percent w/w of the at least one plasticizer.
  • the pharmaceutical composition is designed to prevent immediate release of the at least one active drug substance after physical tampering.
  • 2009/0123386 (assigned to MW Encap Limited) is directed to an abuse deterrent capsule including at least one modifier selected to prevent abuse.
  • the modifier may have a high melting point or be insoluble in aqueous solvents or ethanol.
  • the high melting point excipient may be Poloxamer 188 or PEG 8000.
  • U.S. 2010/0204259 (assigned to Egalet A/S) is directed to immediate release pharmaceutical compositions that are resistant to abuse by intake of alcohol. The release of the drug substance from the immediate release composition is decreased when the composition is exposed to a dissolution medium that includes ethanol.
  • the compositions may be formulated to include at least one polyglycol and at least one effervescent agent.
  • the present disclosure relates to an immediate release, abuse deterrent capsule including an active substance susceptible to abuse, a first polyethylene glycol (PEG) having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons; and a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons.
  • the ratio of the first PEG to the second PEG is less than about 1 :4 w/w.
  • the first PEG and the second PEG together are at least about 60 wt% of the dosage form.
  • the active substance is hydrocodone bitartrate.
  • the active substance is oxycodone hydrochloride (HC1).
  • the capsule includes a grey dye including FD&C Blue #1 , FD&C Yellow #6, and FD&C Red #40.
  • the dye reduces abuse by providing a visual deterrent to injecting.
  • about 60%, 70% 75%, 80%, 85% or about 90% or more of the capsule fill contents are soluble in both water and/or alcohol, e.g., ethanol.
  • the ratio of the first PEG to the second PEG is between about 1 :7 w/w and about 1 : 1 1 w/w.
  • the first PEG has an average molecular weight of about 35,000 Daltons and the second PEG has an average molecular weight of about 3350 Daltons.
  • the capsule includes at least about 2.5 wt% of the active substance.
  • the capsule may be prepared by filling a capsule body with a heated homogenized suspension including the active substance, the first PEG and the second PEG.
  • the present disclosure also relates to an immediate release, abuse deterrent capsule including an active substance susceptible to abuse and polyethylene glycol with a weighted average molecular weight between about 6200 Daltons and about 7800 Daltons.
  • the capsule includes at least about 60 wt% of PEG.
  • the active substance is hydrocodone bitartrate.
  • the active substance is oxycodone HC1.
  • the present disclosure also relates to an immediate release, abuse deterrent capsule including an active substance susceptible to abuse, a first PEG having a melting point greater than or equal to about 60 °C, and a second PEG having a melting point less than or equal to about 57 °C.
  • the contents of the capsule can be solid at 40 °C / 75% relative humidity.
  • at least 90% of the active ingredient can be released from the capsule within 30 minutes following administration or via dissolution testing.
  • at least 75% of the active ingredients can be released from the capsule within 45 minutes following administration or via dissolution testing.
  • the first PEG and the second PEG together are at least about 60 wt% of the capsule.
  • the active substance is hydrocodone bitartrate.
  • the active substance is oxycodone HC1.
  • the present disclosure also relates to a process for the production of an immediate release, abuse deterrent capsule including at least one active substance susceptible to abuse including preparing a homogenized suspension of the at least one active substance susceptible to abuse, a first PEG having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons, and a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons.
  • the process can further include filling the homogenized suspension into a capsule body to produce an encapsulated dosage form.
  • the ratio of the first PEG to the second PEG can be less than about 1 :4 w/w, e.g., between about 1 :7 w/w and about 1 : 1 1 w/w.
  • the first PEG and the second PEG together can be at least about 60 wt% of the capsule.
  • the active substance is hydrocodone bitartrate.
  • the active substance is oxycodone HC1.
  • the capsule can be formed by joining a capsule body with a capsule cap.
  • the present disclosure also relates to a method of treating pain including
  • Figure 1 shows cross sections of a capsule filling machine including the body segment, the cap disc, the hopper, the pumping box, the substation roller, and capsule bodies.
  • Figure 2A shows solutions of grey dye before filtering.
  • Figure 2B shows solutions of grey dye after filtering.
  • Figure 3 shows a summary of an exemplary manufacturing process for formulations of the present disclosure.
  • Figure 4 shows unfiltered solutions of the dosage forms in 190 proof ethanol after shaking at 250 rpm for 3 hours.
  • Figure 5 shows syringe-filtered solutions of the dosage forms in 190 proof ethanol after shaking at 250 rpm for 3 hours.
  • agonist/antagonist combinations aversion, delivery system, prodrug, or a combination of the aforementioned.
  • the categories are: [0020] Physical/Chemical barriers - Physical barriers can prevent chewing, pulverizing, cutting, grating, or grinding. Chemical barriers can resist extraction of the opioid using common solvents like water, alcohol, or other organic solvents. Physical and chemical barriers can change the physical form of an oral drug rendering it less amenable to abuse.
  • An opioid antagonist can be added to interfere with, reduce, or defeat the euphoria associated with abuse.
  • the antagonist can be sequestered and released only upon manipulation of the product.
  • a drug product may be formulated such that the substance that acts as an antagonist is not clinically active when the product is swallowed but becomes active if the product is crushed and injected or snorted.
  • Aversion - Substances can be combined to produce an unpleasant effect if the dosage form is manipulated prior to ingestion or a higher dosage than directed is used.
  • Delivery System including depot injectable formulations and implants
  • Certain drug release designs or the method of drug delivery can offer resistance to abuse.
  • a sustained-release depot injectable formulation that is administered intramuscularly or a subcutaneous implant can be more difficult to manipulate.
  • Prodrug - A prodrug that lacks opioid activity until transformed in the gastrointestinal tract can be unattractive for intravenous injection or intranasal routes of abuse.
  • Combination - Two or more of the above methods can be combined to deter abuse.
  • An opioid analgesic submitted for abuse deterrent formulation (ADF) labeling must show conformance to one or more of these categories.
  • the present disclosure relates to an abuse deterrent dosage form for oral administration, which provides immediate release of an active pharmaceutical substance and conforms to one or more of these categories.
  • the abuse deterrent dosage form of the present disclosure conforms to at least one of the six FDA categories.
  • the abuse deterrent dosage form of the present disclosure conforms to at least two of the six FDA categories.
  • the abuse deterrent dosage form of the present disclosure conforms to at least three of the six FDA categories.
  • the abuse deterrent dosage form of the present disclosure conforms to at least four of the six FDA categories.
  • an abuse deterrent dosage form of the present disclosure conforms to at least five of the six FDA categories.
  • an abuse deterrent dosage form of the present disclosure can reduce abuse by the incorporation of at least one physical barrier.
  • the physical barrier is designed to prevent abuse based on chewing, pulverizing, cutting, grating or grinding.
  • the physical barrier prevents or reduces the effectiveness of these methods.
  • the phrase "abuse deterrent" means that the active substance cannot readily be separated from the formulation in a form suitable for abuse by such means as, for example, grinding.
  • the abuse deterrent form of the present disclosure cannot be easily ground, extracted from, or both. Abuse deterrent measures render it difficult to transform the dosage form into a residue or extract for non-oral administration, such as intranasal or intravenous.
  • the present disclosure relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse, a first PEG having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons, and a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons.
  • the ratio of the first PEG to the second PEG can be less than about 1 :4 w/w.
  • the wt% of active substance in the formulation may also vary depending on the active substance of the dosage form.
  • the dosage form includes at least about 0.1 wt%, 0.2 wt%, 0.3 wt%, 0.4 wt%, 0.5 wt%, 0.6 wt%, 0.7 wt%, 0.8 wt%, 0.9 wt%, 1.0 wt%, 1.1 wt%, 1.2 wt%, 1 .3 wt%, 1.4 wt%, 1.5 wt %, 2 wt%, 2.5 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 7.5 wt%, 8 wt%, 9 wt%, 10 wt%, 1 1 wt%, 12 wt%, 13 wt%, 14 wt%, 15 wt%, 16 wt%, 17 wt%, 18 wt%, 19 wt%, 20 wt%, 21 wt%, 22 .
  • the amount of active substance in the dosage form may range from about 0.10 wt% to about 60 wt%.
  • the amount of active substance in the dosage form may range from about 0.1 wt% to about 1.5 wt%, from about 5 wt% to about 30 wt%, from about 15 wt% to about 20 wt%, from about 15 wt% to about 30 wt%, from about 40 wt% to about 60 wt%, from about 40 wt% to about 50 wt%, or from about 42 wt% to about 46 wt%.
  • the dosage form may be a 100 mg capsule including about 5 mg, about 10 mg, about 15 mg, about 20 mg, or about 30 mg of active substance (e.g., oxycodone HC1).
  • the dosage form may be a 150 mg capsule including about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, or about 45 mg of active substance (e.g., oxycodone HC1).
  • the dosage form may be a 200 mg capsule including about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, or about 60 mg of active substance (e.g., oxycodone HC1).
  • the dosage form may be a 700 mg capsule including about 2.5 mg, about 5 mg, about 7.5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 45 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg or about 100 mg of an active substance (e.g., hydrocodone bitartrate).
  • an active substance e.g., hydrocodone bitartrate
  • the term "active” or “active substance” or “active substance susceptible to abuse” or “API” means any opioid or opioid related compound subject to potential abuse.
  • the active substance may include, without limitation, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone
  • phenadoxone phenazocine
  • phenomorphan phenoperidine
  • piminodine propiram
  • the active can be oxycodone HC1 or hydrocodone bitartrate.
  • the active substance is not oxymorphone.
  • the active substance can be hydrocodone bitartrate or oxycodone HC1.
  • the dosage form of the present disclosure can be rendered abuse deterrent by incorporating PEG in the dosage form.
  • the PEG can deter abuse by preventing at least 50%, or at least 75%, of the capsule weight from being ground to a particle size below 500 ⁇ m, such as after 30 seconds of milling at 10,000 RPM.
  • PEG can also prevent extraction of the active substance from the dosage form using an alcohol.
  • Abusers can use the partial solubility characteristics of dosage form excipients to extract the active substance using alcohol and subsequently burn off the alcohol to form a purer residue containing the active substance.
  • PEG poly(ethylene glycol)
  • Addition of a dye to the dosage form can also result in a colored solution after extraction of the active substance, deterring intravenous injection.
  • PEG in the dosage form can result in the inability of the dosage form, e.g., capsule, to be abused by pulverizing and snorting, pulverizing and injecting, or
  • the abuse deterrent dosage form of the present disclosure may be incapable of being significantly pulverized by physical or mechanical force due at least in part to the waxy characteristics of the PEG.
  • One of the most common means of abuse of an orally administered opioid analgesic involves the manipulation of the oral dosage form in order to cause rapid delivery to the bloodstream via nasal insufflation.
  • the original dosage form In order for insufflation to be used as an effective means of abuse, the original dosage form must be manipulated so as to decrease the particle size of the ingested drug to about 500 ⁇ m or less.
  • a particle size of about 500 ⁇ m or less is necessary for effective intranasal absorption to occur.
  • one way to prevent abuse by nasal insufflation is by capturing the active substance susceptible to abuse in a matrix which is resistant to being physically broken down to produce particles smaller than about 500 ⁇ m.
  • the dosage form of the present disclosure can inhibit manipulation by grinding or pulverizing using common equipment, such as a coffee grinder.
  • the formulation can deter abuse by limiting the particle size to which the formulation may be ground.
  • the formulation prevents the dosage form, or at least substantial portions of the dosage from, from being ground in particles having a particle size of about 500 ⁇ m or less that may pass through the mucus membranes of the nasal cavity.
  • the dosage form can also significantly limit the extraction of the active substance by common solvents (e.g., cold water or distilled aqueous ethanol) from the formulation.
  • the formulation deters abuse by limiting the ability of persons to extract the active substance from the formulation (either intentionally or unintentionally), such that the active substance cannot easily be concentrated for parenteral administration.
  • the abuse deterrent dosage form may also include, but does not require, the incorporation of other deterrents such as antagonists or irritants.
  • the abuse deterrent can work as follows. If the dosage form is extracted with alcohol or an aqueous solution, the PEG and/or dye will also be extracted and cannot easily be separated from the active substance, preventing the preparation of pure drug for intravenous administration. Extraction with a solution would result in a grey/black liquid containing the PEG, dye and active substance.
  • the inclusion of PEG in the formulation can prevent or reduce extraction because PEG can melt and form a wax before the alcohol can be completely evaporated or flashed off, an abuser may not be able to obtain a residue containing the active substance.
  • These properties can allow for an oral drug delivery system that satisfies at least one of the categories in the FDA guidance (e.g., "physical and chemical barriers can change the physical form of an oral drug rendering it less amenable to abuse").
  • the PEG can be capable of allowing immediate release of the active substance, providing abuse deterrence, and/or ensuring the formation of a solid dosage form that is stable at elevated temperatures, for example 40 °C.
  • the PEG provides all three.
  • the dosage form of the present disclosure can accomplish the above capabilities by using a mixture of PEG molecules of at least two different average molecular weights.
  • the dosage form may include a first PEG having an average molecular weight between about 30,000 Daltons and 40,000 Daltons, and a second PEG having an average molecular weight about 3000 Daltons and 4000 Daltons.
  • the first PEG has an average molecular weight of about 20, 000, 21 ,000, 22,000, 23,000, 24,000, 25,000, 26,000, 27,000, 28,000, 29,000, 30,000, 30,500, 31 ,000, 31 ,500, 32,000, 32,500, 33,000, 33,500, 34,000, 34,500, 35,000, 35,500, 36,000, 36,500, 37,000, 37,500, 38,000, 38,500, 39,000, 39,500 or 40,000 Daltons. Any of these values may be used to define a range for the average molecular weight of the first PEG.
  • the first PEG can have an average molecular weight between about 31 ,000 Daltons and about 39,000 Daltons, between about 32,000 Daltons and about 38,000 Daltons, between about 33,000 Daltons and about 37,000 Daltons, between about 34,000 Daltons and about 36,000 Daltons, between about 30,000 Daltons and about 32,000 Daltons, between about 32,000 Daltons and about 34,000 Daltons, between about 36,000 Daltons and about 38,000 Daltons, or between about 38,000 Daltons and about 40,000 Daltons.
  • the second PEG can have an average molecular weight of 3000, 3050, 3100, 3150, 3200, 3250, 3300, 3350, 3400, 3450, 3500, 3550, 3600, 3650, 3700, 3750, 3800, 3850, 3900, 3950 or 4000 Daltons. Any of these values may be used to define a range for the average molecular weight of the second PEG.
  • the second PEG can have an average molecular weight between about 3100 Daltons and about 3900 Daltons, between about 3200 Daltons and about 3800 Daltons, between about 3300 Daltons and about 3700 Daltons, between about 3400 Daltons and about 3600 Daltons, between about 3000 Daltons and 3200 Daltons, between about 3200 Daltons and about 3400 Daltons, between about 3600 Daltons and about 3800 Daltons, or between about 3800 Daltons and about 4000 Daltons.
  • the ratio of the first PEG to the second PEG can be about 3:1, 2:1, 1:1, 1:2, 1:3.1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1 : 19 or 1 :20. Any of these values may be used to define a range for the ratio of the first PEG to the second PEG.
  • the ratio of the first PEG to the second PEG can be between about 1 :2 w/w and about 2:1 w/w, between about 1 :3 w/w and about 1:1 w/w, between about 1 :2 w/w and about 1 : 1 w/w, between about 1 : 1 w/w and about 2: 1 w/w, between about 1 : 1 w/w and about 3 : 1 w/w, between about 1 :4 w/w and about 1:10 w/w, between about 1 :7 w/w/ and about 1:11 w/w, or between about 1 :8 w/w and about 1:10 w/w.
  • the ratio of the first PEG to the second PEG can be less than about 3:1,2:1, 1:1, 1:2, 1:3.1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, l:19or 1 :20.
  • a ratio of 1 :10 is less than a ratio of 1 :9.
  • the total wt% of PEG in the dosage form may vary depending on the active substance, stability, and release profile.
  • the first PEG and the second PEG together are at least about 5 wt%, 10 wt%, 15 wt%, 20 wt%, 25 wt%, 30 wt%, 35 wt%, 36 wt%, 37 wt%, 38 wt%, 39 wt%, 40 wt%, 41 wt%, 42 wt%, 43 wt%, 44 wt%, 45 wt%, 46 wt%, 47 wt%, 48 wt%, 49 wt%, 50 wt%, 51 wt%, 52 wt%, 53 wt%, 54 wt%, 55 wt%, 56 wt%, 57 wt%, 58 wt%, 59 wt%, 60 wt%, 61 wt%, 62
  • the formulation includes a disintegrant.
  • a disintegrant promotes disintegration of the capsule, and dissolution of the active substance, after administration and upon contact with water.
  • the disintegrant may be selected from sodium starch glycolate, cross- linked polyvinylpyrrolidone (e.g. crospovidone), cross-linked sodium carboxymethylcellulose (e.g. croscarmellose sodium) sodium bicarbonate/citric acid, alginic acid or combinations thereof.
  • the disintegrant is selected from sodium starch glycolate, crospovidone and croscarmellose.
  • the dosage form may contain about 1 wt%, 2 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 8 wt%, 9 wt%, 10 wt%, 1 1 wt%, 12 wt%, 13 wt%, 14 wt%, 15 wt%, 16 wt%, 17 wt%, 18 wt%, 19 wt% or 20 wt% of disintegrant. Any of these values may be used to define a range for the wt% of disintegrant.
  • the dosage form may contain between about 1.0 wt% and about 20 wt% of disintegrant.
  • the formulation may contain between about 1.0 wt% and about 10 wt% disintegrant or between about 5 wt% and about 8 wt% disintegrant.
  • the dosage form includes 5 wt% sodium starch glycolate, 8 wt% sodium starch glycolate, 5 wt% crospovidone, or 5 wt% croscarmellose sodium.
  • the dosage form of the present disclosure excludes a
  • the formulation includes a dye.
  • a dye can be useful in deterring abuse by discouraging the abuser from intravenous injection. For example, extraction of the dye along with the active ingredient would result in a colored solution that would discourage the abuser from intravenous injection. Thus, in certain embodiments, the dye reduces abuse by extracting and injecting.
  • the dye may be selected from known dyes suitable for use in pharmaceutical formulations or approved by the FDA for such use. For example, the dye may be FD&C Blue No. 2 or a 50/50 wt% solution of FD&C Blue No. 2 in PEG. In another
  • the dye may be a grey dye including FD&C Blue #1 , FD&C Yellow #6, and FD&C Red #40.
  • the dye may be in a 90% PEG 3350 blend.
  • 14 mg of dye blend can be used in each capsule or about 1.4 mg of concentrated dye.
  • a grey dye is used since it is visually deterring and non-transparent.
  • the dosage form may include about 0.10 wt%, 0.20 wt%, 0.30 wt%, 0.40 wt%, 0.50 wt%, 1 wt%, 2 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 8 wt%, 9 wt%, 10 wt%, 1 1 wt%, 12 wt%, 13 wt%, 14 wt%, 15 wt%, 16 wt%, 17 wt%, 18 wt%, 19 wt%, or 20 wt% dye. Any of these values may be used to define a range for the wt% of the dye.
  • the dosage form may contain between about 0.10 wt% and about 15 wt% dye.
  • the dosage form may contain between about 0.20 wt% and about 1.5 wt% dye, about 0.50 wt% and about 1.0 wt% dye , or about 7 to about 14 wt% dye.
  • the dosage form may include about 1 mg, 1.4 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 nig, 10 mg, 1 1 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg or 30 mg of dye.
  • the dosage form of the present disclosure excludes a dye.
  • the dosage form includes a first dye and a second dye, wherein the first dye has a high solubility in aqueous solution that is higher than the solubility of the second dye in aqueous solution.
  • the first dye has a solubility in aqueous solution of about 1 g, 5 g, 10 g, 30 g, 50 g, 100 g or 500 g in 1 L of aqueous solution and the second dye has a solubility in aqueous solution of about 1 mg, 5 mg, 10 mg, 30 mg, 50 mg, 100 mg, 500 mg, 1 g, or 10 g in 1 L of aqueous solution.
  • the second dye has a high solubility in non-aqueous solution that is greater than the solubility of the first dye in non-aqueous solution.
  • the first dye has a solubility in non-aqueous solution of about 1 mg, 5 mg, 10 mg, 30 mg, 50 mg, 100 mg, 500 mg, 1 g, or 10 g in 1 L of non-aqueous solution
  • the second dye has a solubility in non-aqueous solution of about 1 g, 5 g, 10 g, 30 g, 50 g, 100 g or 500 g in 1 L of non-aqueous solution.
  • the color of the first dye is substantially the same as the color of the second dye.
  • the color of the first dye is substantially different from the color of the second dye.
  • a dye is considered to be soluble in a solvent if about 1 g of the dye can be dissolved in about 10-30 mL of the solvent.
  • a dye is considered to be water soluble if about 1 g of the dye can be dissolved in 10-30 mL of water.
  • the dosage form includes a preservative or antioxidant. The preservative or antioxidant can reduce or limit the degradation or deterioration of the abuse deterrent dosage form.
  • the components of the oral drug delivery system may undergo degradation (e.g., oxidative reduction, chain cleavage) due to oxidation.
  • Preventing degradation can help maintain the abuse deterrent properties of the formulation.
  • the molecular weight of PEG in the formulation affects the resistance to grinding, for example, with a coffee grinder.
  • the addition of a preservative or antioxidant in the formulation that reduces or eliminates the degradation of the molecular weight of PEG may be useful in maintaining the abuse deterrence properties of the dosage form.
  • the addition of a preservative or antioxidant in the dosage form may be necessary to prevent premature degradation of the active substance over the shelf life of the dosage form.
  • the preservative or antioxidant may be selected from preservatives or antioxidants known to one skilled in the art for use in pharmaceutical formulations, such as citric acid, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), erythorbic acid, hypophosphorous acid, lactobionic acid, monothioglycerol, potassium metabisulfite, propyl gallate, racemethionine, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, sodium sulfite, sodium thiosulfate, stannous chloride, sulfur dioxide and tocopherols.
  • preservatives or antioxidants known to one skilled in the art for use in pharmaceutical formulations, such as citric acid, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), erythorbic acid, hypo
  • the formulation, or dosage form may contain between about 0.1 wt% and about 2.0 wt%, or about 0.25 wt% and about 0.75 wt% of preservative or antioxidant.
  • the dosage form of the present disclosure excludes a preservative or antioxidant.
  • the dosage form includes one or more excipients that form a gel in the presence of an alcohol.
  • the alcohol gelling/thickening agent reduces or limits the potential for abuse by preventing extraction of the active substance from the dosage form.
  • the components of the dosage form e.g., active substances, PEG
  • the alcohol gelling/thickening agent does not form a gel in the presence of water.
  • the dosage form can contain up to about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 1 1%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39% or about 40%.
  • These values can be used to define a range, such as about 0.1 wt% to about 40 wt% alcoholic gelling/thickening agent.
  • the dosage form of the present disclosure does not contain an alcohol gelling/thickening agent.
  • the alcohol gelling/thickening agent may be a gelling or thickening agent known to one skilled in the art for use in pharmaceutical formulations, such as acacia, alginic acid, bentonite, calcium acetate, carbomers, carboxymethylcellulose, ethylcellulose, gelatin, hydroxyethylcellulose, hydroxypropyl cellulose, magnesium aluminum silicate, methylcellulose, poloxamers, polyvinyl alcohol, polyvinyl acetate, polyvinylpyrrolidone, sodium alginate, sorbitol derivatives, tragacanth, or xanthan gum.
  • acacia alginic acid
  • bentonite calcium acetate
  • carbomers carboxymethylcellulose
  • ethylcellulose gelatin
  • hydroxyethylcellulose hydroxypropyl cellulose
  • magnesium aluminum silicate magnesium aluminum silicate
  • methylcellulose poloxamers
  • polyvinyl alcohol polyvinyl acetate, polyvinylpyrrolidon
  • the dosage form may additionally include at least one additive independently selected from surfactants, bulking agents, lubricants, flavorings or combination thereof.
  • the abuse deterrent dosage form of the present disclosure is capable of immediate release of the active substance.
  • the dosage form may be manufactured to provide a composition exhibiting an immediate release profile of at least one active substance.
  • immediate release refers to a dosage form that releases the active substance or a
  • the amount of active substance released from the dosage form, e.g., oxycodone HC1 or hydrocodone bitartrate, by exposure to deaerated water within 45 minutes is greater than or equal to 75%.
  • the amount of active substance released from the dosage form, e.g., hydrocodone bitartrate, by exposure to a 0.1 N hydrochloric acid solution within 30 minutes is greater than or equal to 90%.
  • the amount of active substance released from the dosage form, e.g., oxycodone HC1, within 45 minutes is greater than or equal to 75%.
  • the dosage form of the present disclosure releases greater than or equal to about 75% of the active substance within 45 minutes after administration or via dissolution testing. Particularly, the dosage form releases greater than or equal to about 80%, about 85%, about 90%, or about 95% of the active substance within 45 minutes after administration or via dissolution testing.
  • the dosage form of the present disclosure releases greater than or equal to about 90% of the active substance within 30 minutes after administration or via dissolution testing. Particularly, the dosage form releases greater than or equal to about 92%, about 94%, about 96%, or about 98% of the active substance within 30 minutes after
  • the present disclosure also relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse and PEG with a weighted average molecular weight between about 6200 Daltons and about 7800 Daltons.
  • dosage forms containing an average molecular weight of PEG in this particular range have several desirable characteristics including immediate release of the active substance, stability at high temperature conditions (e.g., 40 °C with 75% relative humidity), relatively low viscosity at elevated temperatures (e.g., a viscosity less than or equal to 2000 cP at 75 °C), and/or a relatively high particle size after grinding (e.g., greater than or equal to 50% of the particles having a diameter greater than or equal to 500 ⁇ m after grinding, such as for 30 seconds at 10,000 RPM).
  • Dosage forms including PEG with an average molecular weight between about 6200 Daltons and about 7800 Daltons may be prepared by combining two or more PEGs with different molecular weights.
  • any of the PEGs described herein e.g., PEG 3350 and PEG 35000
  • PEG 3350 and PEG 35000 may be combined to prepare a dosage form including PEG with an average molecular weight range between about 6200 Daltons and about 7800 Daltons.
  • the dosage form includes PEG, or two or more PEGs, with an average molecular weight of about 5000, 5015, 5100, 5200, 5300, 5400, 5500, 5600, 5700, 5800, 5900, 6000, 6100, 6200, 6300, 6400, 6500, 6515, 6600, 6700, 6800, 6900, 7000, 7100, 7200, 7300, 7400, 7500, 7600, 7700, 7800, 7900, 8000, 8100, 8200, 8300, 8400, 8500, 8600, 8700, 8800, 8900, 9000, 9100, 9200, 9300, 9400, 9500, 9600, 9700, 9800, 9900, 10,000, 10,100, 10,200, 10,300, 10,400, 10,500, 10,600, 10,700, 10,800, 10,900, 1 1 ,000, 1 1 ,100, 1 1,200, 1 1 ,300, 1 1 ,400, 1 1 ,500, 1 1 1 1 ,000, 1 ,
  • the dosage form includes PEG, or PEGs, with an average molecular weight between about 6200 Daltons and about 6515 Daltons, between about 6515 Daltons and about 6800 Daltons, or between about 6200 Daltons and about 6800 Daltons.
  • the present disclosure relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse, a first PEG having a melting point greater than or equal to about 60 °C, and a second PEG having a melting point less than or equal to about 57 °C.
  • the dosage form can be a solid at 40 °C / 75% relative humidity, and at least 90% of the active ingredient can be released from the dosage form within 30 minutes following administration or via dissolution testing.
  • the dosage form can be a solid at 40 °C / 75% relative humidity, and at least 75% of the active ingredient can be released from the dosage form within 45 minutes following administration or via dissolution testing.
  • the melting point of PEG can be positively con-elated with molecular weight, i.e. higher molecular weight PEGs have higher melting points.
  • PEGs with an average molecular weight up to 400 Daltons can be considered nonvolatile liquids at room temperature.
  • PEG 600 for example, has a melting range of about 17 to 22 °C, and may be liquid at room temperature but waxy at lower temperatures.
  • PEGs with an average molecular weight of 800 to 2000 Daltons can be considered waxy materials at room temperature with a relatively low melting range.
  • PEG 1500 has a melting point of about 42-46 °C.
  • PEGs with an average molecular weight above 3000 can be considered solids.
  • PEG 3350 has a melting point of about 53-57 °C
  • PEG 35,000 has a melting point of about 60-65 °C.
  • a dosage form with several desirable properties can be formed, including immediate release of an active substance, stability at high temperatures (e.g., 40° C with 75% relative humidity), relatively low viscosity at elevated temperatures (e.g., less than or equal to 2000 cP at 75 °C), and/or a relatively high particle size after grinding (e.g., greater than or equal to 50% of the particles having a diameter greater than or equal to 500 ⁇ m) and/or the incorporation of a chemical barrier which makes it difficult to separate the active substance from the rest of the formulation.
  • stability at high temperatures e.g., 40° C with 75% relative humidity
  • relatively low viscosity at elevated temperatures e.g., less than or equal to 2000 cP at 75 °C
  • a relatively high particle size after grinding e.g., greater than or equal to 50% of the particles having a
  • the dosage form includes a first PEG having a melting temperature greater than or equal to about 52 °C, 53 °C, 54 °C, 55 °C, 56 °C, 57 °C, 58 °C, 59 °C, 60 °C, 61 °C, 62 °C, 63 °C, 64 °C, 65 °C, 66 °C, 67 °C, 68 °C, 69 °C, or 70 °C. Any of these values may be used to define a range of melting temperatures for the first PEG depending on the application.
  • the dosage form may include a first PEG having a melting
  • the dosage form includes a second PEG having a melting temperature less than or equal to about 5 °C, 10 °C, 15 °C, 16 °C, 17 °C, 18 °C, 19 °C, 20 °C, 21 °C, 22 °C, 23 °C, 24 °C, 25 °C, 25 °C, 27 °C, 28 °C, 29 °C, 30 °C, 31 °C, 32 °C, 33 °C, 34 °C, 35 °C, 36 °C, 37 °C, 38 °C, 39 °C, 40 °C, 41 °C, 42 °C, 43 °C, 44 °C, 45 °C, 46 °C, 47 °C, 48 °C, 49 °C, 50 °C, 51 °C, 52 °C, 53 °C, 54 °C, 55 °C, 55 °C, 56 °C, or
  • the dosage form may include a second PEG having a melting temperature between about 17 °C and about 22 °C, between about 42 °C and about 46 °C, between about 53 °C and about 57 °C, or between about 42 °C and about 57 °C.
  • the dosage form includes a first PEG and a second PEG, wherein the first PEG and the second PEG combined have a melting temperature of about 42 °C, 43 °C, 44 °C, 45 °C, 46 °C, 47 °C, 48 °C, 49 °C, 50 °C, 51 °C, 52 °C, 53 °C, 54 °C, 55 °C, 56 °C, 57 °C, 58 °C, 59 °C, 60 °C, 61 °C, 62 °C, 63 °C, 64 °C, 65 °C, 66 °C, 67 °C, 68 °C, 69 °C, 70 °C. Any of these values may be used to define a range of melting temperatures for the combined first and second PEG depending on the application.
  • the first PEG and the second PEG combined may have a melting temperature between about 53 °C and about 65
  • the present disclosure relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse, a first PEG having a melting point greater than or equal to about 60 °C and a second PEG having a viscosity at 100 °C less than or equal to about 1 10 cSt.
  • the dosage form can be a solid at 40 °C / 75% relative humidity, and at least 75% of the active ingredient can be released from the dosage form within 45 minutes following administration or via dissolution testing or at least 90% of the active ingredient can be released from the dosage form within 30 minutes following administration or via dissolution testing.
  • the dosage form includes a second PEG having a viscosity at 100 °C of less than or equal to about 500 cSt, 450 cSt, 400 cSt, 350 cSt, 300 cSt, 250 cSt, 200 cSt, 190 cSt, 180 cSt, 170 cSt, 160 cSt, 158 cSt, 150 cSt, 140 cSt, 130 cSt, 123 cSt, 120 cSt, 1 10 cSt, 105 cSt, 100 cSt, 99 cSt, 93 cSt, 90 cSt, 87 cSt, 80 cSt, 76 cSt, 75 cSt, 73 cSt, 70 cSt, 67 cSt, 60 cSt, 50 cSt, 49 cSt, 48 cSt, 47
  • the dosage form may include a second PEG having a viscosity between about 4.0 cSt and about 49.0 cSt, between about 16.0 cSt and about 49.0 cSt, between about 25.0 cSt and about 32.0 cSt, or between about 76 cSt and about 1 10 cSt.
  • the formulation of the present disclosure can have a viscosity at 100 °C of about 40 cSt, 41 cSt, 42 cSt, 43 cSt, 44 cSt, 45 cSt, 46 cSt, 47 cSt, 48 cSt, 49 cSt, 50 cSt, 51 cSt, 52 cSt, 53 cSt, 54 cSt, 55 cSt, 56 cSt, 57 cSt, 58 cSt, 59 cSt, 60 cSt, 61 cSt, 62 cSt, 63 cSt, 64 cSt, 65 cSt, 66 cSt, 67 cSt, 68 cSt, 69 cSt, 70 cSt, 71 cSt, 72 cSt, 73 cSt, 74 cSt, 75
  • the formulation of the present disclosure may have a viscosity between about 500 cSt and about 2000 cSt, or between about 800 cSt and about 1900 cSt.
  • the formulation or dosage form is a solid at room temperature and/or at 100 °C and has not measureable viscosity.
  • the present disclosure relates to a process for the production of an oral, immediate release, abuse deterrent dosage form including preparing a homogenized suspension of at least one active substance susceptible to abuse, a first PEG, and a second PEG.
  • the first PEG can have an average molecular weight between about 30,000 Daltons and about 40,000 Daltons
  • the second PEG can have an average molecular weight between about 3000 Daltons and about 4000 Daltons.
  • the ratio of the first PEG to the second PEG can be less than about 1 :4 w/w.
  • the process can further include dispensing or filling a homogenized suspension into a capsule to produce the dosage form.
  • the capsule can be formed by joining a capsule body with a capsule cap.
  • the first PEG and the second PEG together may be any wt% of the dosage form as described herein, for example at least about 60 wt% of the dosage form.
  • the active substance is hydrocodone bitartrate.
  • the active substance is oxycodone HC1.
  • the abuse deterrent dosage forms of the present disclosure are capsules.
  • the abuse deterrent dosage forms of the present disclosure may be produced by liquid filled encapsulation.
  • Liquid filled encapsulation is a process in which active pharmaceutical ingredients are suspended or emulsified in a carrier matrix and filled into capsules.
  • the capsules are usually made of hard gelatin or hydroxypropyl methylcellulose.
  • the internal solid phase API e.g., oxycodone HC1 or hydrocodone bitartrate
  • PEGs with average molecular weights greater than about 1500 Daltons are ideal for liquid filled capsules because they are thermoplastics that melt at temperatures below the melting point of the hard gelatin capsule ( ⁇ 70 °C ) and are solids at room temperature. If the filling material is liquid at room temperature, a banding process can be used. This process adds a gelatin band around the point where the capsule body and cap join to create a unified capsule body to prevent leakage.
  • the formulation of the present disclosure can include a band.
  • the liquid fill process can begin by dispensing excipients (e.g., PEG and stabilizers/preservatives) and API according to theoretical percent weights of the final capsule fill weight.
  • excipients e.g., PEG and stabilizers/preservatives
  • API e.g., PEG and stabilizers/preservatives
  • API e.g., PEG and stabilizers/preservatives
  • API e.g., PEG and stabilizers/preservatives
  • the suspension can be pumped through jacketed hoses (to maintain the internal kettle temperature to prevent solidification in the hose) to a hopper on the capsule filling machine.
  • An illustration of a capsule filling machine is provided in Figure 1.
  • the capsule filling hopper can also be jacketed to heat the suspension to prevent solidification.
  • the capsule filling machine can contain a separate hopper which operators fill with hard gelatin capsules.
  • the hopper can feed into a rectifying drum which can align all capsules in the same direction. Once aligned, the capsules can sit vertically in a cap disk which can allow for separation of the body and cap via vacuum.
  • a positive displacement piston pump can be used to draw the product in from the jacketed hopper and dispense the suspension into the capsule body through a set of changeable nozzles. Fill weight adjustment can be achieved by varying the piston stroke of the pump. These changes can be made throughout the process due to frequent in-process capsule weight checks.
  • the capsule body and cap can be joined via pusher pins which raise the capsule body upwards and into the capsule cap, which are held in place above the capsule body by a joining block.
  • the pusher pins can then push the unified capsule out of the cap disk and discharge them from the machine.
  • the capsules can then be allowed to cool at room temperature on trays and can be each weight checked via a capsule weigh checking machine. Following this, the capsules can then be placed into a final output drum.
  • Automatic capsule filling machines can have the ability to produce 500 to 150,000 capsules an hour with a very high degree of accuracy.
  • the present disclosure relates to a dosage form as described herein prepared by filling a capsule body with a heated homogenized suspension including an active substance, a first PEG and a second PEG.
  • the homogenized suspension including an active substance, a first PEG, and a second PEG melts at a temperature of about 42 °C, 43 °C, 44 °C, 45 °C, 46 °C, 47 °C, 48 °C, 49 °C, 50 °C, 51 °C, 52 °C, 53 °C, 54 °C, 55 °C, 56 °C, 57 °C, 58 °C, 59 °C, 60 °C, 61 °C, 62 °C, 63 °C, 64 °C, 65 °C, 66 °C, 67 °C, 68 °C, 69 °C, 70 °C, 71 °C, 72 °
  • the homogenized suspension has a melting temperature between about 53 °C and about 65 °C.
  • the homogenized suspension including an active substance, a first PEG and a second PEG melts at temperatures below 77 °C, i.e., the melting point of the hard gelatin capsule.
  • the present disclosure relates to a method of treating pain including administering to an individual in need thereof a therapeutically effective amount of a dosage form as described herein.
  • the dosage form can be used for the management of moderate to severe pain where the use of an opioid analgesic is appropriate.
  • the dosage form can provide rapid onset of analgesia for the treatment of moderate to severe pain.
  • the dosage form e.g., a hard gelatin capsule, can be administered orally every 4-6 hours as needed.
  • the disclosures of all cited references including publications, patents, and patent applications are expressly incorporated herein by reference in their entirety. Further, when an amount, concentration, or other value or parameter is given as either a range, preferred range, or a list of upper preferable values and lower preferable values, this is to be understood as specifically disclosing all ranges formed from any pair of any upper range limit or preferred value and any lower range limit or preferred value, regardless of whether ranges are separately disclosed.
  • a formulation was prepared containing 30 mg APAP and a 50:50 ratio of PEG 3350:1500 g/mol and 0.50% FD&C dye in size 3 white opaque capsules.
  • Three capsule fill weights were evaluated: 100 mg, 150 mg, and 200 mg. These formulations were tested for dissolution.
  • Table 1 below list the dissociation data of size 3 capsules containing 30 mg APAP, a 50:50 ratio of PEG 3350:1500 g/mol, and 0.5% FD&C dye.
  • These dosage forms contain water- and ethanol-soluble FD&C dyes, e.g., 0.5% FD&C dye, to deter extraction of the API and intravenous injection of the solution. Further rendering of the drug solution would be required to separate the pure API from the PEG and FD&C dyes.
  • PEG 1450 (NF grade available from Dow Chemical Company) can be used in place of PEG 1500 in the oxycodone HC1 dosage forms. Additional exemplary oxycodone HC1 dosage forms are shown in Table 2 below. 1 % citric acid may be used in the dosage forms as an API stabilizer.
  • ADF oxycodone HC1 liquid fill capsule formulations containing varying amounts of PEG 1450 and PEG 35000 were evaluated.
  • Acetaminophen (APAP) was used as a tracer drug for oxycodone HC1.
  • the formulations are shown in Table 3 below.
  • the target amount of APAP was 30 mg per capsule, and the target fill weight was 100 mg (batch number 92) or 200 mg (batch numbers 93-94).
  • the capsules contained 30% w/w (batch number 92) or 15% w/w (batch numbers 93-94) APAP. Size 3 opaque hard gelatin capsules were used.
  • a formulation is considered to deter intranasal abuse if ⁇ 75% of the particles are ⁇ 500 ⁇ m in diameter after grinding. As shown in Table 3, the percentage of particles ⁇ 500 ⁇ m in diameter after grinding ranged from 90% to 92%. Thus all of the oxycodone formulations met the standard of ⁇ 75% of the particles being ⁇ 500 ⁇ m in diameter after grinding.
  • the oxycodone HC1 formulations were also analyzed to determine melting temperature.
  • the capsules were held at 40 °C / 75% relative humidity for 72 hours.
  • the batch number 92 and 93 formulations containing 100% and 82% PEG 35000, respectively, were solid at these conditions, while the batch number 94 formulation containing 59% PEG 35000 had a much softer fill.
  • hot melt fill capsules are sufficiently viscous at elevated temperatures to allow for flow of the fill into the capsules.
  • additional oxycodone HC1 formulations containing PEG 35000 and either PEG 3350 or PEG 1450 were evaluated by measuring viscosity at 75 °C at 50 rpm.
  • Formulations were weighed out according to total wt % of a 15g batch. Each formulation was poured into a viscosity testing crucible and placed in an 80 °C water bath to melt.
  • the formulations were mixed using a stainless steel spatula and transferred to a Brookfield DV-II+ Pro Viscometer (VIS29 NCD: Upon Use) utilizing Spindle: S27 (Small Sample Adapter).
  • the viscometer was equipped with a water jacketed crucible platform. Once the melt temperature reached 75 °C, a viscosity reading was taken in centipoise (cP). Based on manufacturer specifications, an acceptable viscosity for the purposes of this study is ⁇ 1000 cP.
  • the particle size after grinding and stability at 40 °C / 75% relative humidity (RH) was also determined. For the grinding analysis, an acceptable particle size after grinding was considered to be ⁇ 75% particles ⁇ 500 ⁇ m in diameter. All
  • formulations were size 3 opaque hard gelatin capsules.
  • PEG 1450 had a viscosity of 1288 cP at 75 °C / 50 rpm, above the manufacturer specification of ⁇ 1000 cP. Accordingly, viscosity was not measured for the formulations containing higher percentages of PEG 35000 (i.e. batch numbers 97-99). In addition, batch number 100 was not sufficiently stable for storage, since this formulation was a very viscous liquid at the stability test conditions of 40 °C / 75% RH.
  • Formulations containing PEG 35000 and PEG 3350 were also evaluated. As shown in Table 6 below, the formulation containing 6.97% PEG 35000 and 62.7% PEG 3350 (batch number 104) and the formulation containing 7.7% PEG 35000 and 69.3% PEG 3350 (batch number 105) met all of the criteria for particle size after grinding, viscosity, and stability.
  • Varying concentrations of FD&C Blue #2, green (FD&C Blue #2 and FD&C Yellow #5), FD&C Yellow #5, FD&C Red #40, and grey dye (FD&C Blue#l, FD&C Yellow #6, FD&C Red #40) were evaluated by dissolving them in a 95% ethanol 5% purified water (190 proof) solution and passing the solution through a syringe filter. After syringe filtering the dye solutions were visually evaluated for color intensity and rated on a scale of 0 to 5, with 0 indicating no color and 5 indicating dark, significant color. As shown in the Table 7 below, the blue and green dyes exhibited the highest color intensity at low concentrations, e.g. 0.25% w/w. Solutions of grey dye before and after filtering are shown in Figures 2A and 2B, respectively. The grey dye was particularly striking and less appealing. An acceptable color scale designation after extraction of the dye is ⁇ 4 on a scale of 1 to 5, with 5 being the highest level of color.
  • the dye can be grey. Grey can be chosen because it is darker than the others and can be effective at a lower relative concentration. Grey dye can allow for the most visually deterring form with the least amount of dye present in the formulation.
  • Abusers of opioid products often adulterate the product to promote more rapid release of the active ingredient.
  • the products can be chewed and swallowed, crushed and inhaled, or extracted in water or alcohol (either crushed or intact) to produce a solution that can be used for intravenous administration or dried for insufflation of a purified product.
  • Adulteration of the products can enable a more rapid delivery of active than can be achieved by ingestion of the intact product. This rapid onset, high exposure is associated with euphoria, drug liking, and greater abuse potential.
  • All C-II narcotic drug products tested can be cut with an edged surface (e.g., scissors or a razor blade) and therefore can potentially be abused, with forces that are substantially lower than what has been reported using the breaking strength test or equivalent (e.g., >500 N). Flattening the tablets using forces greater than 500 N (with traditional "tablet breaking force" definitions) does not address abuse deterrence potential in the tested C-II narcotic drug products.
  • the formulation of the present disclosure can be resistant to abuse by nasal insufflation or extraction due to, in part, the waxy nature of the formulation contents and the solubility of the excipients.
  • the excipients can be both water and alcohol soluble to create a formulation that makes it time consuming and costly to extract oxycodone HC1 from the formulation contents without also extracting the excipients.
  • a high molecular weight PEG can be included because of its solubility properties (e.g., soluble in both alcohol and water) and its resistance to grinding to particle sizes of less than 500 ⁇ m.
  • High-molecular weight PEGs are less viscous at melt temperatures than long chain PEO molecules and are soluble in both water and alcohol.
  • Dyes can also be used and chosen to be soluble in both water and alcohol to produce a dark colored solution upon extraction and filtering as a visual deterrent to abuse.
  • the formulation can include the following components listed in the Table 8 below, including a number of different dyes. Table 8 below lists the components along with their solubility information taken from the various literature sources and tested experimentally (e.g., 200 proof ethanol and filtered through a 0.45 micrometer PTFE filter). The extraction of the active to a pure form can be very difficult using water or alcohol.
  • a conventional tablet or powder-filled capsule can be easily crushed to create a fine powder.
  • the waxy material contained in the formulations of the present disclosure can make it difficult to manipulate into particles small enough to be easily absorbed by the nasal mucosa.
  • the waxy material may also congeal once introduced to the semi-aqueous environment of the nasal passages, which can make it difficult to introduce the oxycodone HC1 or hydrocodone bitartrate to the bloodstream via the nasal passages.
  • the formulations of the present disclosure can contain one or more of the following barriers to abuse.
  • Insufflation The formulation can be formulated to resist grinding to particle sizes of less than about 500 ⁇ m.
  • Extraction and Purification The formulation can be formulated with water- and alcohol-soluble dyes to create a dark colored solution upon extraction that can be visually unappealing to intravenous drug users.
  • the water- and alcohol- soluble excipients can present obstacles to purification of the active. In some formulations, if the solvent is flashed off or otherwise evaporated, the excipients can return to the same waxy, dark- colored form as before being introduced to the solvent.
  • Vaporization The formulation can contain an active, such as oxycodone HC1, which can degrade at temperatures close to where vaporization occurs. Chewing - Because the formulation is an immediate release formulation, it is not expected that crushing or cutting the dosage form will result in an especially rapid release of the drug to produce a "euphoric high.”
  • Table 9 lists exemplary formulations for the oxycodone HC1 abuse deterrent formulation capsules.
  • Formulations of the present disclosure were manufactured by the following exemplary process.
  • the components of the hot-melt suspension consisting of Polyethylene Glycol 3350, Polyethylene Glycol 35000, Dye Blend, Grey Powder, Citric Acid and Oxycodone HC1 were dispensed according to theoretical batch quantities based on formulation weight percents.
  • Polyethylene Glycol 3350, Polyethylene Glycol 35000, Dye Blend, Grey Powder, Oxycodone HC1 and Citric Acid were added to an Olsa 150 Liter Kettle and heated to a temperature of 70 ⁇ 20 °C. Utilizing the homogenizer mixer, external anchor blades and internal mixing blades, the melt was then mixed until uniform
  • the formulations of the present disclosure are stable upon storage at 25, 30, 35, 40 or 45 °C, and at 60%, 65%, 70% or 75% relative humidity, e.g., 30 °C / 65% RH or 40 °C / 75% RH.
  • the formulation of the present disclosure can be stable under any of these conditions for up to 1 , 2, 3, 4, 5, 6, 9, 12, 16, 18, 24, or 36 months.
  • Cutting the dosage form can be performed in order to increase the surface area of the product prior to ingesting it in an effort to increase the rate of dissolution into the digestive tract.
  • Cutting can also be used to increase the efficiency of grinding or extraction.
  • Grinding the dosage form can be performed in order to decrease the particle size of the product more efficiently than cutting in an effort to insufflate (snort) for immediate release into the blood vessels of the nasal passages.
  • a readily available tool used for grinding is a commercially available coffee grinder.
  • a drug product is considered abuse deterrent if the % material in the pan ( ⁇ 500 ⁇ m) is ⁇ 50%.
  • a dosage form which, when ground, produces ⁇ 50% of the material on a per-dosage form basis available for nasal insufflation ( ⁇ 500 ⁇ m) is considered abuse deterrent.
  • Texture analysis is the mechanical testing of pharmaceutical products in order to measure their physical properties.
  • the Retsch Knife Mill GRINDOMIX GM200 (TE96) was utilized to mimic a commercially available coffee grinder (Mr. Coffee) in order to grind the drug products into a particle size that is suitable for intranasal abuse (insufflation).
  • the Retsch Knife Mill GRINDOMIX GM200 utilizes a circular blade attachment to mimic commercially available coffee grinders.
  • the GM200 has a top speed of 10,000 revolutions per minute (rpm), while commercially available coffee grinders have a top speed of approximately 20,000 rpm (an approximate two-fold increase in speed when comparing the GM200 to a Mr. Coffee grinder).
  • the approximate two-fold increase in blade diameter (1 18 mm vs. 60 mm, when comparing the GM200 to a Mr. Coffee grinder, respectively) compensates for the approximate twofold decrease in top speed via the inversely proportional relationship of the two variables.
  • the torque provided by the GM200 is significantly higher than the torque provided by a Mr.
  • Coffee grinder (0.860 Nm (Newton meters) of the GM200 vs. 0.062 Nm of the Mr. Coffee grinder, respectively), which additionally illustrates the ability (or lack thereof) of the Mr. Coffee grinder to modify the drug products into a particle size suitable for intranasal abuse.
  • the weight of the 35 mesh sieve and sample pan was recorded.
  • the testing apparatus was assembled with the 35 mesh sieve above the sample pan.
  • the composite sample was transferred to the testing apparatus and analyzed utilizing the following parameters: 1 minute analysis time and no pulse.
  • the analyzed 35 mesh sieve and sample pan were weighed.
  • the % material remaining on the 35 mesh sieve ( ⁇ 500 ⁇ m) and in the sample pan ( ⁇ 500 ⁇ m) was calculated using the following equation: x 100
  • Table 10 shows the particle size after grinding for the oxycodone HC1 formulations tested. During testing it was observed by visual observation that the capsule portion of the dosage form of all evaluated batches was not being significantly modified by TE96, and that the majority of the capsule portion remained in the 35 mesh sieve ( ⁇ 500 ⁇ m). The grinding / particle size analysis for this protocol is based on weight differences, which, when the capsule portion is taken into account, can skew the results towards a higher proportion of particles ⁇ 500 ⁇ m.
  • the average percentage of particles ⁇ 500 ⁇ m after grinding for the oxycodone HC1 capsules ranged from 62.3% to 68.2%.
  • approximately 20% of particles by weight of an Immediate Release (IR) Roxicodone® formulation were ⁇ 500 ⁇ m after the same grinding procedure.
  • Table 13 summarizes the grinding results and statistical analysis of the % material ⁇ 500 ⁇ m for the Present Disclosure 15mg and Roxicodone® 15mg tablets (Mallinckrodt).
  • Another method of rending a drug product abusable is via extraction of the active substance from the dosage form to produce a pure residue.
  • This method can be performed, and is often performed, using a high proof alcohol or an aqueous media.
  • the formulation of the present disclosure can be readily soluble in both aqueous and alcohol environments when the contents are removed from the capsule. Therefore, aqueous and alcohol extraction techniques were evaluated. Solutions were analyzed qualitatively for solution color following filtration, as well as quantitatively for % label claim (LC) (with regards to oxycodone HCI) of solution following filtration. Additionally, evaporated residual samples were analyzed qualitatively for residue color following evaporation, as well as quantitatively for purity determination following the %LC calculations.
  • LC % label claim
  • the quantitative results of the analysis determine the % purity (with regards to oxycodone HCI) of the extracted sample solution described above.
  • a drug product can be considered abuse deterrent if the % residue purity is ⁇ 50%. In other embodiments, less than or equal to 40%, 45%, 55%, 60%, 65%, 70% or 75%. Residue purity levels (with regards to the API) ⁇ 50% can infer that the excipient load is greater than the API level contained in the residue. In one embodiment, this can be considered abuse deterrent with regards to potential intravenous abuse of a purified residue.
  • the F-test and t-tests was analyzed in order to determine if the drug products provide statistically different % purity values.
  • Tables 14 and 15 show the formulation of the present disclosure results in 9% and 9% purity with regards to oxycodone HC1, in alcohol and aqueous environments, respectively. This is in comparison to Roxicodone® 15mg, which has a purity of 68% and 19% purity in alcohol and aqueous environments, respectively. This data proves the formulation of the present disclosure is statistically different than Roxicodone 18 in both alcohol and aqueous extracts.
  • Color is one identifying characteristic of commercial drug products. Color can be applied to the dosage form in two ways: dye or coating.
  • High potency alcohol i.e., ⁇ 190 proof (95%)
  • Dyes or coatings can potentially be used to alter the physical appearance of the extracted solution of drug product (i.e., turn the resulting solution a noticeable color).
  • the inclusion of one or more dyes in a drug formulation is one method to render a formulation abuse deterrent.
  • Significant discoloration of an extraction product from a formulation subject to abuse can discourage a potential abuser from using (e.g., injecting or ingesting) the extraction product.

Abstract

The present disclosure relates to an oral, immediate release, abuse deterrent liquid filled capsule containing polyethylene glycol and at feast one active pharmaceutical ingredient susceptible to abuse. The dosage form is abuse deterrent to parenteral administration. The present disclosure also relates to processes of preparing the dosage form.

Description

IMMEDIATE RELEASE ABUSE DETERRENT LIQUID FILL DOSAGE FORM
Cross Reference to Related Applications
[0001 ] This application claims priority to U.S. Provisional Application No. 62/025,878 filed July 17, 2014, the disclosure of which is incorporated herein by reference in their entirety.
Field of the Technology
[0002] The present disclosure relates to an oral immediate release, abuse deterrent dosage form. The dosage form contains polyethylene glycol (PEG) to reduce abuse by non-oral administration routes, e.g. intranasal and/or intravenous. The composition of PEG is designed to allow for immediate release of the active ingredient while deterring abuse and maintaining stability of the dosage form at elevated temperatures.
Background
[0003] FDA-approved drugs are provided in many different forms based on the type of active substance, the indication treated and the preferred route of administration. These forms include enteral formulations (e.g., tablets, capsules or pills), parenteral formulations (e.g., injectable formulations such as intravenous, subcutaneous, intramuscular and intraarticular), liquid formulations (e.g., elixirs), lyophilized formulations and topical formulations. A majority of the FDA-approved drugs are currently available in enteral form, as either a tablet or capsule.
[0004] Several formulations have been investigated for deterring abuse, either by oral ingestion of the drug with alcohol, or by non-oral administration routes such as intranasal and/or intravenous administration. For example, U.S. 2014/0010873 (assigned to Egalet Ltd.) is directed to an abuse-deterrent pharmaceutical composition including at least one polyethylene oxide and at least one plasticizer. The polyethylene oxide has an average molecular weight of at least 1 ,000,000 Daltons, and the pharmaceutical composition includes at least 5 percent w/w of the at least one plasticizer. The pharmaceutical composition is designed to prevent immediate release of the at least one active drug substance after physical tampering. U.S. 2009/0123386 (assigned to MW Encap Limited) is directed to an abuse deterrent capsule including at least one modifier selected to prevent abuse. The modifier may have a high melting point or be insoluble in aqueous solvents or ethanol. For example, the high melting point excipient may be Poloxamer 188 or PEG 8000. U.S. 2010/0204259 (assigned to Egalet A/S) is directed to immediate release pharmaceutical compositions that are resistant to abuse by intake of alcohol. The release of the drug substance from the immediate release composition is decreased when the composition is exposed to a dissolution medium that includes ethanol. The compositions may be formulated to include at least one polyglycol and at least one effervescent agent.
Summary
[0005] The present disclosure relates to an immediate release, abuse deterrent capsule including an active substance susceptible to abuse, a first polyethylene glycol (PEG) having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons; and a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons. The ratio of the first PEG to the second PEG is less than about 1 :4 w/w.
[0006] In some embodiments, the first PEG and the second PEG together are at least about 60 wt% of the dosage form. In some embodiments, the active substance is hydrocodone bitartrate. In other embodiments, the active substance is oxycodone hydrochloride (HC1). In some embodiments the capsule includes a grey dye including FD&C Blue #1 , FD&C Yellow #6, and FD&C Red #40. In certain embodiments, the dye reduces abuse by providing a visual deterrent to injecting. In certain embodiments, about 60%, 70% 75%, 80%, 85% or about 90% or more of the capsule fill contents are soluble in both water and/or alcohol, e.g., ethanol. In certain embodiments, the ratio of the first PEG to the second PEG is between about 1 :7 w/w and about 1 : 1 1 w/w. In some embodiments, the first PEG has an average molecular weight of about 35,000 Daltons and the second PEG has an average molecular weight of about 3350 Daltons. In some embodiments, the capsule includes at least about 2.5 wt% of the active substance. The capsule may be prepared by filling a capsule body with a heated homogenized suspension including the active substance, the first PEG and the second PEG.
[0007] The present disclosure also relates to an immediate release, abuse deterrent capsule including an active substance susceptible to abuse and polyethylene glycol with a weighted average molecular weight between about 6200 Daltons and about 7800 Daltons. In certain embodiments, the capsule includes at least about 60 wt% of PEG. In some embodiments, the active substance is hydrocodone bitartrate. In other embodiments, the active substance is oxycodone HC1.
[0008] The present disclosure also relates to an immediate release, abuse deterrent capsule including an active substance susceptible to abuse, a first PEG having a melting point greater than or equal to about 60 °C, and a second PEG having a melting point less than or equal to about 57 °C. The contents of the capsule can be solid at 40 °C / 75% relative humidity. In some embodiments, at least 90% of the active ingredient can be released from the capsule within 30 minutes following administration or via dissolution testing. In other embodiments, at least 75% of the active ingredients can be released from the capsule within 45 minutes following administration or via dissolution testing. In some embodiments, the first PEG and the second PEG together are at least about 60 wt% of the capsule. In particular embodiments, the active substance is hydrocodone bitartrate. In other embodiments, the active substance is oxycodone HC1.
[0009] The present disclosure also relates to a process for the production of an immediate release, abuse deterrent capsule including at least one active substance susceptible to abuse including preparing a homogenized suspension of the at least one active substance susceptible to abuse, a first PEG having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons, and a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons. The process can further include filling the homogenized suspension into a capsule body to produce an encapsulated dosage form. The ratio of the first PEG to the second PEG can be less than about 1 :4 w/w, e.g., between about 1 :7 w/w and about 1 : 1 1 w/w.
[0010] In certain embodiments of the aforementioned process, the first PEG and the second PEG together can be at least about 60 wt% of the capsule. In particular embodiments, the active substance is hydrocodone bitartrate. In other embodiments, the active substance is oxycodone HC1. In certain embodiments the capsule can be formed by joining a capsule body with a capsule cap.
[001 1 ] The present disclosure also relates to a method of treating pain including
administering to a subject in need thereof a therapeutically effective amount of any of the aforementioned capsules. Brief Description of the Drawings
[0012] The foregoing and other advantages provided by the present disclosure will be more fully understood from the following description of exemplary embodiments when read together with the accompanying drawings, in which:
[0013] Figure 1 shows cross sections of a capsule filling machine including the body segment, the cap disc, the hopper, the pumping box, the substation roller, and capsule bodies.
[0014] Figure 2A shows solutions of grey dye before filtering. Figure 2B shows solutions of grey dye after filtering.
[0015] Figure 3 shows a summary of an exemplary manufacturing process for formulations of the present disclosure.
[0016] Figure 4 shows unfiltered solutions of the dosage forms in 190 proof ethanol after shaking at 250 rpm for 3 hours.
[0017] Figure 5 shows syringe-filtered solutions of the dosage forms in 190 proof ethanol after shaking at 250 rpm for 3 hours.
Detailed Description
[0018] Abuse of prescription drugs, particularly opioids, is a serious and growing public health concern. To address this concern, new formulations are being developed that contain abuse-deterrent properties. Abuse deterrent properties include properties that make product manipulation more difficult or make abuse of the manipulated product less attractive or rewarding.
[0019] Recently the FDA issued a draft guidance for industry related to formulations having abuse deterrent properties. Guidance for Industry: Abuse -Deterrent Opioids - Evaluation and Labeling, U.S. Department of Health and Human Services, FDA, CDER, January 2013, the entire contents of which are incorporated herein by reference. These guidelines separate abuse deterrent formulations into six categories, including: physical/chemical barriers,
agonist/antagonist combinations, aversion, delivery system, prodrug, or a combination of the aforementioned. As described by the FDA guidance, the categories are: [0020] Physical/Chemical barriers - Physical barriers can prevent chewing, pulverizing, cutting, grating, or grinding. Chemical barriers can resist extraction of the opioid using common solvents like water, alcohol, or other organic solvents. Physical and chemical barriers can change the physical form of an oral drug rendering it less amenable to abuse.
[0021 ] Agonist/Antagonist combinations - An opioid antagonist can be added to interfere with, reduce, or defeat the euphoria associated with abuse. The antagonist can be sequestered and released only upon manipulation of the product. For example, a drug product may be formulated such that the substance that acts as an antagonist is not clinically active when the product is swallowed but becomes active if the product is crushed and injected or snorted.
[0022] Aversion - Substances can be combined to produce an unpleasant effect if the dosage form is manipulated prior to ingestion or a higher dosage than directed is used.
[0023] Delivery System (including depot injectable formulations and implants) - Certain drug release designs or the method of drug delivery can offer resistance to abuse. For example, a sustained-release depot injectable formulation that is administered intramuscularly or a subcutaneous implant can be more difficult to manipulate.
[0024] Prodrug - A prodrug that lacks opioid activity until transformed in the gastrointestinal tract can be unattractive for intravenous injection or intranasal routes of abuse.
[0025] Combination - Two or more of the above methods can be combined to deter abuse.
[0026] An opioid analgesic submitted for abuse deterrent formulation (ADF) labeling must show conformance to one or more of these categories. The present disclosure relates to an abuse deterrent dosage form for oral administration, which provides immediate release of an active pharmaceutical substance and conforms to one or more of these categories. In one embodiment, the abuse deterrent dosage form of the present disclosure conforms to at least one of the six FDA categories. In another embodiment, the abuse deterrent dosage form of the present disclosure conforms to at least two of the six FDA categories. In another embodiment, the abuse deterrent dosage form of the present disclosure conforms to at least three of the six FDA categories. In another embodiment, the abuse deterrent dosage form of the present disclosure conforms to at least four of the six FDA categories. In another embodiment, the abuse deterrent dosage form of the present disclosure conforms to at least five of the six FDA categories. [0027] For example, an abuse deterrent dosage form of the present disclosure can reduce abuse by the incorporation of at least one physical barrier. The physical barrier is designed to prevent abuse based on chewing, pulverizing, cutting, grating or grinding. Preferably, the physical barrier prevents or reduces the effectiveness of these methods. As used herein, the phrase "abuse deterrent" means that the active substance cannot readily be separated from the formulation in a form suitable for abuse by such means as, for example, grinding. The abuse deterrent form of the present disclosure cannot be easily ground, extracted from, or both. Abuse deterrent measures render it difficult to transform the dosage form into a residue or extract for non-oral administration, such as intranasal or intravenous.
[0028] In one embodiment, the present disclosure relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse, a first PEG having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons, and a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons. The ratio of the first PEG to the second PEG can be less than about 1 :4 w/w. The wt% of active substance in the formulation may also vary depending on the active substance of the dosage form. In some embodiments, the dosage form includes at least about 0.1 wt%, 0.2 wt%, 0.3 wt%, 0.4 wt%, 0.5 wt%, 0.6 wt%, 0.7 wt%, 0.8 wt%, 0.9 wt%, 1.0 wt%, 1.1 wt%, 1.2 wt%, 1 .3 wt%, 1.4 wt%, 1.5 wt %, 2 wt%, 2.5 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 7.5 wt%, 8 wt%, 9 wt%, 10 wt%, 1 1 wt%, 12 wt%, 13 wt%, 14 wt%, 15 wt%, 16 wt%, 17 wt%, 18 wt%, 19 wt%, 20 wt%, 21 wt%, 22 wt%, 23 wt%, 24 wt%, 25 wt%, 26 wt%, 27 wt%, 28 wt%, 29 wt%, 30 wt%, 31 wt%, 32 wt%, 33 wt%, 34 wt%, 35 wt%, 36 wt%, 37 wt%, 38 wt%, 39 wt%, 40 wt%, 41 wt%, 42 wt%, 43 wt%, 44 wt%, 45 wt%, 46 wt%, 47 wt%, 48 wt%, 49 wt%, 50 wt%, 51 wt%, 52 wt%, 53 wt%, 54 wt%, 55 wt%, 56 wt%, 57 wt%, 58 wt%, 59 wt%, 60 wt%, 65 wt%, 69 wt%, 70 wt%, 75 wt%, 80 wt%, 85 wt%, 88 wt%, 90 wt%, or 95 wt% of the active substance. Any of these values may be used to define a range for the wt% of the active substance depending on the application. For example, the amount of active substance in the dosage form may range from about 0.10 wt% to about 60 wt%. Particularly, the amount of active substance in the dosage form may range from about 0.1 wt% to about 1.5 wt%, from about 5 wt% to about 30 wt%, from about 15 wt% to about 20 wt%, from about 15 wt% to about 30 wt%, from about 40 wt% to about 60 wt%, from about 40 wt% to about 50 wt%, or from about 42 wt% to about 46 wt%. [0029] For example, the dosage form may be a 100 mg capsule including about 5 mg, about 10 mg, about 15 mg, about 20 mg, or about 30 mg of active substance (e.g., oxycodone HC1). In other embodiments, the dosage form may be a 150 mg capsule including about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, or about 45 mg of active substance (e.g., oxycodone HC1). In other embodiments, the dosage form may be a 200 mg capsule including about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, or about 60 mg of active substance (e.g., oxycodone HC1). In other embodiments, the dosage form may be a 700 mg capsule including about 2.5 mg, about 5 mg, about 7.5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 45 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg or about 100 mg of an active substance (e.g., hydrocodone bitartrate).
[0030] As used herein, the term "active" or "active substance" or "active substance susceptible to abuse" or "API" means any opioid or opioid related compound subject to potential abuse. The active substance may include, without limitation, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levophenacylmorphan, levorphanol, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbulphine, narceine, nicomorphine, norpipanone, opium, oxycodone, papvretum, pentazocine,
phenadoxone, phenazocine, phenomorphan, phenoperidine, piminodine, propiram,
propoxyphene, sufentanil, tilidine, tapentadol, and tramadol, and pharmaceutically acceptable salts and mixtures thereof. For example, in some embodiments the active can be oxycodone HC1 or hydrocodone bitartrate. In the dosage forms of the present disclosure, the active substance is not oxymorphone.
[0031 ] In particular, the active substance can be hydrocodone bitartrate or oxycodone HC1. The dosage form of the present disclosure can be rendered abuse deterrent by incorporating PEG in the dosage form. The PEG can deter abuse by preventing at least 50%, or at least 75%, of the capsule weight from being ground to a particle size below 500 μm, such as after 30 seconds of milling at 10,000 RPM. PEG can also prevent extraction of the active substance from the dosage form using an alcohol. Abusers can use the partial solubility characteristics of dosage form excipients to extract the active substance using alcohol and subsequently burn off the alcohol to form a purer residue containing the active substance. The inclusion of PEG in the formulation can prevent or reduce extraction because PEG can melt and form a wax before the alcohol can be completely evaporated or flashed off, an abuser may not be able to obtain a residue containing the active substance. Addition of a dye to the dosage form can also result in a colored solution after extraction of the active substance, deterring intravenous injection. By selecting the appropriate average molecular weight and quantity of PEG present within a dosage form, the characteristics of the dosage form can be manipulated in a way to create a wide array of abuse deterrent capsules having immediate release profiles.
[0032] Inclusion of PEG in the dosage form can result in the inability of the dosage form, e.g., capsule, to be abused by pulverizing and snorting, pulverizing and injecting, or
combinations thereof. For example, the abuse deterrent dosage form of the present disclosure may be incapable of being significantly pulverized by physical or mechanical force due at least in part to the waxy characteristics of the PEG.
[0033] One of the most common means of abuse of an orally administered opioid analgesic involves the manipulation of the oral dosage form in order to cause rapid delivery to the bloodstream via nasal insufflation. In order for insufflation to be used as an effective means of abuse, the original dosage form must be manipulated so as to decrease the particle size of the ingested drug to about 500 μm or less. A particle size of about 500 μm or less is necessary for effective intranasal absorption to occur. By limiting the quantity of particles under about 500 μm that an abuser can obtain by reasonable methods, one can render insufflation ineffective as a means of abuse. Thus one way to prevent abuse by nasal insufflation is by capturing the active substance susceptible to abuse in a matrix which is resistant to being physically broken down to produce particles smaller than about 500 μm.
[0034] The dosage form of the present disclosure can inhibit manipulation by grinding or pulverizing using common equipment, such as a coffee grinder. For example, the formulation can deter abuse by limiting the particle size to which the formulation may be ground. The formulation prevents the dosage form, or at least substantial portions of the dosage from, from being ground in particles having a particle size of about 500 μm or less that may pass through the mucus membranes of the nasal cavity. The dosage form can also significantly limit the extraction of the active substance by common solvents (e.g., cold water or distilled aqueous ethanol) from the formulation. For example, the formulation deters abuse by limiting the ability of persons to extract the active substance from the formulation (either intentionally or unintentionally), such that the active substance cannot easily be concentrated for parenteral administration. The abuse deterrent dosage form may also include, but does not require, the incorporation of other deterrents such as antagonists or irritants.
[0035] For example, in one embodiment, the abuse deterrent can work as follows. If the dosage form is extracted with alcohol or an aqueous solution, the PEG and/or dye will also be extracted and cannot easily be separated from the active substance, preventing the preparation of pure drug for intravenous administration. Extraction with a solution would result in a grey/black liquid containing the PEG, dye and active substance. The inclusion of PEG in the formulation can prevent or reduce extraction because PEG can melt and form a wax before the alcohol can be completely evaporated or flashed off, an abuser may not be able to obtain a residue containing the active substance. These properties can allow for an oral drug delivery system that satisfies at least one of the categories in the FDA guidance (e.g., "physical and chemical barriers can change the physical form of an oral drug rendering it less amenable to abuse").
[0036] The PEG can be capable of allowing immediate release of the active substance, providing abuse deterrence, and/or ensuring the formation of a solid dosage form that is stable at elevated temperatures, for example 40 °C. In some embodiments, the PEG provides all three. The dosage form of the present disclosure can accomplish the above capabilities by using a mixture of PEG molecules of at least two different average molecular weights. For example, the dosage form may include a first PEG having an average molecular weight between about 30,000 Daltons and 40,000 Daltons, and a second PEG having an average molecular weight about 3000 Daltons and 4000 Daltons.
[0037] In some embodiments, the first PEG has an average molecular weight of about 20, 000, 21 ,000, 22,000, 23,000, 24,000, 25,000, 26,000, 27,000, 28,000, 29,000, 30,000, 30,500, 31 ,000, 31 ,500, 32,000, 32,500, 33,000, 33,500, 34,000, 34,500, 35,000, 35,500, 36,000, 36,500, 37,000, 37,500, 38,000, 38,500, 39,000, 39,500 or 40,000 Daltons. Any of these values may be used to define a range for the average molecular weight of the first PEG. For example, the first PEG can have an average molecular weight between about 31 ,000 Daltons and about 39,000 Daltons, between about 32,000 Daltons and about 38,000 Daltons, between about 33,000 Daltons and about 37,000 Daltons, between about 34,000 Daltons and about 36,000 Daltons, between about 30,000 Daltons and about 32,000 Daltons, between about 32,000 Daltons and about 34,000 Daltons, between about 36,000 Daltons and about 38,000 Daltons, or between about 38,000 Daltons and about 40,000 Daltons.
[0038] In some embodiments, the second PEG can have an average molecular weight of 3000, 3050, 3100, 3150, 3200, 3250, 3300, 3350, 3400, 3450, 3500, 3550, 3600, 3650, 3700, 3750, 3800, 3850, 3900, 3950 or 4000 Daltons. Any of these values may be used to define a range for the average molecular weight of the second PEG. For example, the second PEG can have an average molecular weight between about 3100 Daltons and about 3900 Daltons, between about 3200 Daltons and about 3800 Daltons, between about 3300 Daltons and about 3700 Daltons, between about 3400 Daltons and about 3600 Daltons, between about 3000 Daltons and 3200 Daltons, between about 3200 Daltons and about 3400 Daltons, between about 3600 Daltons and about 3800 Daltons, or between about 3800 Daltons and about 4000 Daltons.
[0039] In some embodiments, the ratio of the first PEG to the second PEG can be about 3:1, 2:1, 1:1, 1:2, 1:3.1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1 : 19 or 1 :20. Any of these values may be used to define a range for the ratio of the first PEG to the second PEG. For example, in some embodiments, the ratio of the first PEG to the second PEG can be between about 1 :2 w/w and about 2:1 w/w, between about 1 :3 w/w and about 1:1 w/w, between about 1 :2 w/w and about 1 : 1 w/w, between about 1 : 1 w/w and about 2: 1 w/w, between about 1 : 1 w/w and about 3 : 1 w/w, between about 1 :4 w/w and about 1:10 w/w, between about 1 :7 w/w/ and about 1:11 w/w, or between about 1 :8 w/w and about 1:10 w/w. In other embodiments, the ratio of the first PEG to the second PEG can be less than about 3:1,2:1, 1:1, 1:2, 1:3.1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, l:19or 1 :20. For example, a ratio of 1 :10 is less than a ratio of 1 :9.
[0040] The total wt% of PEG in the dosage form may vary depending on the active substance, stability, and release profile. In some embodiments, the first PEG and the second PEG together are at least about 5 wt%, 10 wt%, 15 wt%, 20 wt%, 25 wt%, 30 wt%, 35 wt%, 36 wt%, 37 wt%, 38 wt%, 39 wt%, 40 wt%, 41 wt%, 42 wt%, 43 wt%, 44 wt%, 45 wt%, 46 wt%, 47 wt%, 48 wt%, 49 wt%, 50 wt%, 51 wt%, 52 wt%, 53 wt%, 54 wt%, 55 wt%, 56 wt%, 57 wt%, 58 wt%, 59 wt%, 60 wt%, 61 wt%, 62 wt%, 63 wt%, 64 wt%, 65 wt%, 66 wt%, 67 wt%, 68 wt%, 69 wt%, 69.7 wt%, 70 wt%, 71 wt%, 72 wt%, 73 wt%, 74 wt%, 75 wt%, 76 wt%, 77 wt%, 78 wt%, 79 wt%, 80 wt%, 85 wt%, 88 wt%, 90 wt%, or 95 wt% of the dosage form.
[0041 ] In one embodiment, the formulation includes a disintegrant. A disintegrant promotes disintegration of the capsule, and dissolution of the active substance, after administration and upon contact with water. The disintegrant may be selected from sodium starch glycolate, cross- linked polyvinylpyrrolidone (e.g. crospovidone), cross-linked sodium carboxymethylcellulose (e.g. croscarmellose sodium) sodium bicarbonate/citric acid, alginic acid or combinations thereof. In particular embodiments, the disintegrant is selected from sodium starch glycolate, crospovidone and croscarmellose. The dosage form may contain about 1 wt%, 2 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 8 wt%, 9 wt%, 10 wt%, 1 1 wt%, 12 wt%, 13 wt%, 14 wt%, 15 wt%, 16 wt%, 17 wt%, 18 wt%, 19 wt% or 20 wt% of disintegrant. Any of these values may be used to define a range for the wt% of disintegrant. For example, the dosage form may contain between about 1.0 wt% and about 20 wt% of disintegrant. Particularly, the formulation may contain between about 1.0 wt% and about 10 wt% disintegrant or between about 5 wt% and about 8 wt% disintegrant. In certain embodiments, the dosage form includes 5 wt% sodium starch glycolate, 8 wt% sodium starch glycolate, 5 wt% crospovidone, or 5 wt% croscarmellose sodium. In another embodiment, the dosage form of the present disclosure excludes a
disintegrant.
[0042] In some embodiments, the formulation includes a dye. A dye can be useful in deterring abuse by discouraging the abuser from intravenous injection. For example, extraction of the dye along with the active ingredient would result in a colored solution that would discourage the abuser from intravenous injection. Thus, in certain embodiments, the dye reduces abuse by extracting and injecting. The dye may be selected from known dyes suitable for use in pharmaceutical formulations or approved by the FDA for such use. For example, the dye may be FD&C Blue No. 2 or a 50/50 wt% solution of FD&C Blue No. 2 in PEG. In another
embodiment, the dye may be a grey dye including FD&C Blue #1 , FD&C Yellow #6, and FD&C Red #40. The dye may be in a 90% PEG 3350 blend. In certain embodiments, 14 mg of dye blend can be used in each capsule or about 1.4 mg of concentrated dye. In certain embodiments a grey dye is used since it is visually deterring and non-transparent. The dosage form may include about 0.10 wt%, 0.20 wt%, 0.30 wt%, 0.40 wt%, 0.50 wt%, 1 wt%, 2 wt%, 3 wt%, 4 wt%, 5 wt%, 6 wt%, 7 wt%, 8 wt%, 9 wt%, 10 wt%, 1 1 wt%, 12 wt%, 13 wt%, 14 wt%, 15 wt%, 16 wt%, 17 wt%, 18 wt%, 19 wt%, or 20 wt% dye. Any of these values may be used to define a range for the wt% of the dye. For example, the dosage form may contain between about 0.10 wt% and about 15 wt% dye. Particularly, the dosage form may contain between about 0.20 wt% and about 1.5 wt% dye, about 0.50 wt% and about 1.0 wt% dye , or about 7 to about 14 wt% dye. In certain embodiments, the dosage form may include about 1 mg, 1.4 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 nig, 10 mg, 1 1 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg or 30 mg of dye. In another embodiment, the dosage form of the present disclosure excludes a dye.
[0043] In some embodiments, the dosage form includes a first dye and a second dye, wherein the first dye has a high solubility in aqueous solution that is higher than the solubility of the second dye in aqueous solution. For example, in some embodiments the first dye has a solubility in aqueous solution of about 1 g, 5 g, 10 g, 30 g, 50 g, 100 g or 500 g in 1 L of aqueous solution and the second dye has a solubility in aqueous solution of about 1 mg, 5 mg, 10 mg, 30 mg, 50 mg, 100 mg, 500 mg, 1 g, or 10 g in 1 L of aqueous solution. In some embodiments, the second dye has a high solubility in non-aqueous solution that is greater than the solubility of the first dye in non-aqueous solution. For example, in some embodiments, the first dye has a solubility in non-aqueous solution of about 1 mg, 5 mg, 10 mg, 30 mg, 50 mg, 100 mg, 500 mg, 1 g, or 10 g in 1 L of non-aqueous solution, and the second dye has a solubility in non-aqueous solution of about 1 g, 5 g, 10 g, 30 g, 50 g, 100 g or 500 g in 1 L of non-aqueous solution. In some embodiments, the color of the first dye is substantially the same as the color of the second dye. In other embodiments, the color of the first dye is substantially different from the color of the second dye. For the purposes of the present disclosure, a dye is considered to be soluble in a solvent if about 1 g of the dye can be dissolved in about 10-30 mL of the solvent. For example, a dye is considered to be water soluble if about 1 g of the dye can be dissolved in 10-30 mL of water. [0044] In another embodiment, the dosage form includes a preservative or antioxidant. The preservative or antioxidant can reduce or limit the degradation or deterioration of the abuse deterrent dosage form. For example, the components of the oral drug delivery system (e.g., active substances, PEG) may undergo degradation (e.g., oxidative reduction, chain cleavage) due to oxidation. Preventing degradation can help maintain the abuse deterrent properties of the formulation. For instance, the molecular weight of PEG in the formulation affects the resistance to grinding, for example, with a coffee grinder. The addition of a preservative or antioxidant in the formulation that reduces or eliminates the degradation of the molecular weight of PEG may be useful in maintaining the abuse deterrence properties of the dosage form. In addition to maintaining abuse deterrence, the addition of a preservative or antioxidant in the dosage form may be necessary to prevent premature degradation of the active substance over the shelf life of the dosage form.
[0045] The preservative or antioxidant may be selected from preservatives or antioxidants known to one skilled in the art for use in pharmaceutical formulations, such as citric acid, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), erythorbic acid, hypophosphorous acid, lactobionic acid, monothioglycerol, potassium metabisulfite, propyl gallate, racemethionine, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, sodium sulfite, sodium thiosulfate, stannous chloride, sulfur dioxide and tocopherols. The formulation, or dosage form, may contain between about 0.1 wt% and about 2.0 wt%, or about 0.25 wt% and about 0.75 wt% of preservative or antioxidant. In another embodiment, the dosage form of the present disclosure excludes a preservative or antioxidant.
[0046] In some embodiments, the dosage form includes one or more excipients that form a gel in the presence of an alcohol. The alcohol gelling/thickening agent reduces or limits the potential for abuse by preventing extraction of the active substance from the dosage form. For example, when introduced to an alcohol solution, the components of the dosage form (e.g., active substances, PEG) may become trapped in a gel/viscous liquid which prevents extraction and subsequent alcohol evaporation to produce a pure active substance. In one embodiment, the alcohol gelling/thickening agent does not form a gel in the presence of water. The dosage form can contain up to about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 1 1%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39% or about 40%. These values can be used to define a range, such as about 0.1 wt% to about 40 wt% alcoholic gelling/thickening agent. In another embodiment, the dosage form of the present disclosure does not contain an alcohol gelling/thickening agent.
[0047] The alcohol gelling/thickening agent may be a gelling or thickening agent known to one skilled in the art for use in pharmaceutical formulations, such as acacia, alginic acid, bentonite, calcium acetate, carbomers, carboxymethylcellulose, ethylcellulose, gelatin, hydroxyethylcellulose, hydroxypropyl cellulose, magnesium aluminum silicate, methylcellulose, poloxamers, polyvinyl alcohol, polyvinyl acetate, polyvinylpyrrolidone, sodium alginate, sorbitol derivatives, tragacanth, or xanthan gum.
[0048] The dosage form may additionally include at least one additive independently selected from surfactants, bulking agents, lubricants, flavorings or combination thereof.
[0049] The abuse deterrent dosage form of the present disclosure is capable of immediate release of the active substance. The dosage form may be manufactured to provide a composition exhibiting an immediate release profile of at least one active substance. As used herein,
"immediate release" refers to a dosage form that releases the active substance or a
pharmaceutically acceptable salt thereof, e.g., oxycodone HC1 or hydrocodone bitartrate, substantially completely into the gastrointestinal tract of the user within a period of less than an hour, and often less than about 45 minutes or 30 minutes from ingestion. In one embodiment, the amount of active substance released from the dosage form, e.g., oxycodone HC1 or hydrocodone bitartrate, by exposure to deaerated water within 45 minutes is greater than or equal to 75%. In another embodiment, the amount of active substance released from the dosage form, e.g., hydrocodone bitartrate, by exposure to a 0.1 N hydrochloric acid solution within 30 minutes is greater than or equal to 90%. In other embodiment, the amount of active substance released from the dosage form, e.g., oxycodone HC1, within 45 minutes is greater than or equal to 75%.
[0050] In one embodiment, the dosage form of the present disclosure releases greater than or equal to about 75% of the active substance within 45 minutes after administration or via dissolution testing. Particularly, the dosage form releases greater than or equal to about 80%, about 85%, about 90%, or about 95% of the active substance within 45 minutes after administration or via dissolution testing.
[0051 ] In other embodiments, the dosage form of the present disclosure releases greater than or equal to about 90% of the active substance within 30 minutes after administration or via dissolution testing. Particularly, the dosage form releases greater than or equal to about 92%, about 94%, about 96%, or about 98% of the active substance within 30 minutes after
administration or via dissolution testing.
[0052] The present disclosure also relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse and PEG with a weighted average molecular weight between about 6200 Daltons and about 7800 Daltons. In one embodiment, dosage forms containing an average molecular weight of PEG in this particular range have several desirable characteristics including immediate release of the active substance, stability at high temperature conditions (e.g., 40 °C with 75% relative humidity), relatively low viscosity at elevated temperatures (e.g., a viscosity less than or equal to 2000 cP at 75 °C), and/or a relatively high particle size after grinding (e.g., greater than or equal to 50% of the particles having a diameter greater than or equal to 500 μm after grinding, such as for 30 seconds at 10,000 RPM). Dosage forms including PEG with an average molecular weight between about 6200 Daltons and about 7800 Daltons may be prepared by combining two or more PEGs with different molecular weights. For example, any of the PEGs described herein (e.g., PEG 3350 and PEG 35000) may be combined to prepare a dosage form including PEG with an average molecular weight range between about 6200 Daltons and about 7800 Daltons.
[0053] In particular embodiments, the dosage form includes PEG, or two or more PEGs, with an average molecular weight of about 5000, 5015, 5100, 5200, 5300, 5400, 5500, 5600, 5700, 5800, 5900, 6000, 6100, 6200, 6300, 6400, 6500, 6515, 6600, 6700, 6800, 6900, 7000, 7100, 7200, 7300, 7400, 7500, 7600, 7700, 7800, 7900, 8000, 8100, 8200, 8300, 8400, 8500, 8600, 8700, 8800, 8900, 9000, 9100, 9200, 9300, 9400, 9500, 9600, 9700, 9800, 9900, 10,000, 10,100, 10,200, 10,300, 10,400, 10,500, 10,600, 10,700, 10,800, 10,900, 1 1 ,000, 1 1 ,100, 1 1,200, 1 1 ,300, 1 1 ,400, 1 1 ,500, 1 1 ,600, 1 1 ,675, or 1 1 ,700 Daltons. Any of these values may be used to define a range of average molecular weights for PEG, or PEGs, depending on the application. For example, in some embodiments, the dosage form includes PEG, or PEGs, with an average molecular weight between about 6200 Daltons and about 6515 Daltons, between about 6515 Daltons and about 6800 Daltons, or between about 6200 Daltons and about 6800 Daltons.
[0054] In other embodiments, the present disclosure relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse, a first PEG having a melting point greater than or equal to about 60 °C, and a second PEG having a melting point less than or equal to about 57 °C. The dosage form can be a solid at 40 °C / 75% relative humidity, and at least 90% of the active ingredient can be released from the dosage form within 30 minutes following administration or via dissolution testing. The dosage form can be a solid at 40 °C / 75% relative humidity, and at least 75% of the active ingredient can be released from the dosage form within 45 minutes following administration or via dissolution testing.
[0055] The melting point of PEG can be positively con-elated with molecular weight, i.e. higher molecular weight PEGs have higher melting points. For example, PEGs with an average molecular weight up to 400 Daltons can be considered nonvolatile liquids at room temperature. PEG 600, for example, has a melting range of about 17 to 22 °C, and may be liquid at room temperature but waxy at lower temperatures. PEGs with an average molecular weight of 800 to 2000 Daltons can be considered waxy materials at room temperature with a relatively low melting range. For example, PEG 1500 has a melting point of about 42-46 °C. PEGs with an average molecular weight above 3000 can be considered solids. For example, PEG 3350 has a melting point of about 53-57 °C, and PEG 35,000 has a melting point of about 60-65 °C. By combining a PEG with a relatively low melting point (e.g., PEG 3350) with a PEG with a relatively high melting point (e.g., PEG 35,000) a dosage form with several desirable properties can be formed, including immediate release of an active substance, stability at high temperatures (e.g., 40° C with 75% relative humidity), relatively low viscosity at elevated temperatures (e.g., less than or equal to 2000 cP at 75 °C), and/or a relatively high particle size after grinding (e.g., greater than or equal to 50% of the particles having a diameter greater than or equal to 500 μm) and/or the incorporation of a chemical barrier which makes it difficult to separate the active substance from the rest of the formulation.
[0056] In some embodiments, the dosage form includes a first PEG having a melting temperature greater than or equal to about 52 °C, 53 °C, 54 °C, 55 °C, 56 °C, 57 °C, 58 °C, 59 °C, 60 °C, 61 °C, 62 °C, 63 °C, 64 °C, 65 °C, 66 °C, 67 °C, 68 °C, 69 °C, or 70 °C. Any of these values may be used to define a range of melting temperatures for the first PEG depending on the application. For example, the dosage form may include a first PEG having a melting
temperature from about 52 °C to about 60 °C, from about 55 °C to about 60 °C, from about 53 °C to about 57 °C, from about 53 °C to about 56 °C, from about 55 °C to about 58 °C, from about 60 °C to about 65 °C, or from about 60 °C to about 70 °C.
[0057] In some embodiments, the dosage form includes a second PEG having a melting temperature less than or equal to about 5 °C, 10 °C, 15 °C, 16 °C, 17 °C, 18 °C, 19 °C, 20 °C, 21 °C, 22 °C, 23 °C, 24 °C, 25 °C, 25 °C, 27 °C, 28 °C, 29 °C, 30 °C, 31 °C, 32 °C, 33 °C, 34 °C, 35 °C, 36 °C, 37 °C, 38 °C, 39 °C, 40 °C, 41 °C, 42 °C, 43 °C, 44 °C, 45 °C, 46 °C, 47 °C, 48 °C, 49 °C, 50 °C, 51 °C, 52 °C, 53 °C, 54 °C, 55 °C, 55 °C, 56 °C, or about 57 °C. Any of these values may be used to define a range of melting temperatures for the second PEG depending on the application. For example, the dosage form may include a second PEG having a melting temperature between about 17 °C and about 22 °C, between about 42 °C and about 46 °C, between about 53 °C and about 57 °C, or between about 42 °C and about 57 °C.
[0058] In some embodiments, the dosage form includes a first PEG and a second PEG, wherein the first PEG and the second PEG combined have a melting temperature of about 42 °C, 43 °C, 44 °C, 45 °C, 46 °C, 47 °C, 48 °C, 49 °C, 50 °C, 51 °C, 52 °C, 53 °C, 54 °C, 55 °C, 56 °C, 57 °C, 58 °C, 59 °C, 60 °C, 61 °C, 62 °C, 63 °C, 64 °C, 65 °C, 66 °C, 67 °C, 68 °C, 69 °C, 70 °C. Any of these values may be used to define a range of melting temperatures for the combined first and second PEG depending on the application. For example, the first PEG and the second PEG combined may have a melting temperature between about 53 °C and about 65 °C.
[0059] In other embodiments, the present disclosure relates to an oral, immediate release, abuse deterrent dosage form including an active substance susceptible to abuse, a first PEG having a melting point greater than or equal to about 60 °C and a second PEG having a viscosity at 100 °C less than or equal to about 1 10 cSt. The dosage form can be a solid at 40 °C / 75% relative humidity, and at least 75% of the active ingredient can be released from the dosage form within 45 minutes following administration or via dissolution testing or at least 90% of the active ingredient can be released from the dosage form within 30 minutes following administration or via dissolution testing. [0060] In some embodiments, the dosage form includes a second PEG having a viscosity at 100 °C of less than or equal to about 500 cSt, 450 cSt, 400 cSt, 350 cSt, 300 cSt, 250 cSt, 200 cSt, 190 cSt, 180 cSt, 170 cSt, 160 cSt, 158 cSt, 150 cSt, 140 cSt, 130 cSt, 123 cSt, 120 cSt, 1 10 cSt, 105 cSt, 100 cSt, 99 cSt, 93 cSt, 90 cSt, 87 cSt, 80 cSt, 76 cSt, 75 cSt, 73 cSt, 70 cSt, 67 cSt, 60 cSt, 50 cSt, 49 cSt, 48 cSt, 47 cSt, 46 cSt, 45 cSt, 44 cSt, 43 cSt, 42 cSt, 41 cSt, 40 cSt, 39 cSt, 38 cSt, 37 cSt 36 cSt, 35 cSt, 34 cSt, 33 cSt, 32 cSt, 31 cSt, 30 cSt, 29 cSt, 28 cSt, 27 cSt, 26 cSt, 25 cSt, 24 cSt, 23 cSt, 22 cSt, 21 cSt, 20 cSt, 19 cSt, 18 cSt, 17 cSt, 16 cSt, 15 cSt, 14 cSt, 13 cSt, 12 cSt, 1 1 cSt, 10 cSt, 9 cSt, 8 cSt, 7 cSt, 6 cSt, 5 cSt, or about 4 cSt. Any of these values may be used to define a range of viscosities for the second PEG depending on the application. For example, the dosage form may include a second PEG having a viscosity between about 4.0 cSt and about 49.0 cSt, between about 16.0 cSt and about 49.0 cSt, between about 25.0 cSt and about 32.0 cSt, or between about 76 cSt and about 1 10 cSt.
[0061] In some embodiments, the formulation of the present disclosure can have a viscosity at 100 °C of about 40 cSt, 41 cSt, 42 cSt, 43 cSt, 44 cSt, 45 cSt, 46 cSt, 47 cSt, 48 cSt, 49 cSt, 50 cSt, 51 cSt, 52 cSt, 53 cSt, 54 cSt, 55 cSt, 56 cSt, 57 cSt, 58 cSt, 59 cSt, 60 cSt, 61 cSt, 62 cSt, 63 cSt, 64 cSt, 65 cSt, 66 cSt, 67 cSt, 68 cSt, 69 cSt, 70 cSt, 71 cSt, 72 cSt, 73 cSt, 74 cSt, 75 cSt, 76 cSt, 77cSt, 78 cSt, 80 cSt, 90 cSt, 100 cSt, 1 10 cSt, 120 cSt, 130 cSt, 140 cSt, 150 cSt, 158 cSt, 160 cSt, 170 cSt, 180 cSt, 190 cSt, 200 cSt, 250 cSt, 300 cSt, 350 cSt, 400 cSt, 450 cSt 500 cSt, 600 cSt, 700 cSt, 800 cSt, 900 cSt, 1000 cSt, 1 100 cSt, 1200 cSt, 1300 cSt, 1400 cSt, 1500 cSt, 1600 cSt, 1700 cSt, 1800 cSt, 1900 cSt, or about 2000 cSt. Any of these values may be used to define a range of viscosities for the formulation of the present disclosure depending on the application. For example, the formulation of the present disclosure may have a viscosity between about 500 cSt and about 2000 cSt, or between about 800 cSt and about 1900 cSt. In some embodiments, the formulation or dosage form is a solid at room temperature and/or at 100 °C and has not measureable viscosity.
[0062] In another embodiment, the present disclosure relates to a process for the production of an oral, immediate release, abuse deterrent dosage form including preparing a homogenized suspension of at least one active substance susceptible to abuse, a first PEG, and a second PEG. For example, the first PEG can have an average molecular weight between about 30,000 Daltons and about 40,000 Daltons, and the second PEG can have an average molecular weight between about 3000 Daltons and about 4000 Daltons. The ratio of the first PEG to the second PEG can be less than about 1 :4 w/w. The process can further include dispensing or filling a homogenized suspension into a capsule to produce the dosage form. In some embodiments, the capsule can be formed by joining a capsule body with a capsule cap. The first PEG and the second PEG together may be any wt% of the dosage form as described herein, for example at least about 60 wt% of the dosage form. In some embodiments of the processes described herein, the active substance is hydrocodone bitartrate. In other embodiments, the active substance is oxycodone HC1. In certain embodiments, the abuse deterrent dosage forms of the present disclosure are capsules.
[0063] The abuse deterrent dosage forms of the present disclosure may be produced by liquid filled encapsulation. Liquid filled encapsulation is a process in which active pharmaceutical ingredients are suspended or emulsified in a carrier matrix and filled into capsules. The capsules are usually made of hard gelatin or hydroxypropyl methylcellulose. One of the advantages of this dosage form is that it requires fewer excipients and processing steps than other traditional compressed solid dosage forms. The internal solid phase API (e.g., oxycodone HC1 or hydrocodone bitartrate) can be suspended in a PEG external fluid phase. In one embodiment, PEGs with average molecular weights greater than about 1500 Daltons are ideal for liquid filled capsules because they are thermoplastics that melt at temperatures below the melting point of the hard gelatin capsule (≤70 °C ) and are solids at room temperature. If the filling material is liquid at room temperature, a banding process can be used. This process adds a gelatin band around the point where the capsule body and cap join to create a unified capsule body to prevent leakage. In some embodiments, the formulation of the present disclosure can include a band.
[0064] In one embodiment, the liquid fill process can begin by dispensing excipients (e.g., PEG and stabilizers/preservatives) and API according to theoretical percent weights of the final capsule fill weight. Following this step, the PEG powders or flakes and dyes are pre-melted before they are added to a homogenizing mixing kettle which can maintain the PEG above its melting point via jacketing on the kettle. When the PEG is completely fluid, the API and other non-melting stabilizers and/or preservatives can be mixed in to form a homogenized suspension. This can occur with the aid of mechanical agitation by way of several internal stirring arms. Once a homogenized suspension is attained (in some embodiments newer kettles can be equipped with NIR probes to indicate when this happens), the suspension can be pumped through jacketed hoses (to maintain the internal kettle temperature to prevent solidification in the hose) to a hopper on the capsule filling machine. An illustration of a capsule filling machine is provided in Figure 1.
[0065] The capsule filling hopper can also be jacketed to heat the suspension to prevent solidification. The capsule filling machine can contain a separate hopper which operators fill with hard gelatin capsules. The hopper can feed into a rectifying drum which can align all capsules in the same direction. Once aligned, the capsules can sit vertically in a cap disk which can allow for separation of the body and cap via vacuum. To fill the capsule, a positive displacement piston pump can be used to draw the product in from the jacketed hopper and dispense the suspension into the capsule body through a set of changeable nozzles. Fill weight adjustment can be achieved by varying the piston stroke of the pump. These changes can be made throughout the process due to frequent in-process capsule weight checks.
[0066] Once the capsule body is filled, the capsule body and cap can be joined via pusher pins which raise the capsule body upwards and into the capsule cap, which are held in place above the capsule body by a joining block. The pusher pins can then push the unified capsule out of the cap disk and discharge them from the machine. The capsules can then be allowed to cool at room temperature on trays and can be each weight checked via a capsule weigh checking machine. Following this, the capsules can then be placed into a final output drum. Automatic capsule filling machines can have the ability to produce 500 to 150,000 capsules an hour with a very high degree of accuracy.
[0067] In some embodiments, the present disclosure relates to a dosage form as described herein prepared by filling a capsule body with a heated homogenized suspension including an active substance, a first PEG and a second PEG. In some embodiments, the homogenized suspension including an active substance, a first PEG, and a second PEG melts at a temperature of about 42 °C, 43 °C, 44 °C, 45 °C, 46 °C, 47 °C, 48 °C, 49 °C, 50 °C, 51 °C, 52 °C, 53 °C, 54 °C, 55 °C, 56 °C, 57 °C, 58 °C, 59 °C, 60 °C, 61 °C, 62 °C, 63 °C, 64 °C, 65 °C, 66 °C, 67 °C, 68 °C, 69 °C, 70 °C, 71 °C, 72 °C, 73 °C, 74 °C, or 75 °C. Any of these values may be used to define a range of melting temperatures for the homogenized suspension. For example, in certain embodiments, the homogenized suspension has a melting temperature between about 53 °C and about 65 °C. In particular embodiments, the homogenized suspension including an active substance, a first PEG and a second PEG melts at temperatures below 77 °C, i.e., the melting point of the hard gelatin capsule. In another embodiment, the present disclosure relates to a method of treating pain including administering to an individual in need thereof a therapeutically effective amount of a dosage form as described herein. The dosage form can be used for the management of moderate to severe pain where the use of an opioid analgesic is appropriate. The dosage form can provide rapid onset of analgesia for the treatment of moderate to severe pain. The dosage form, e.g., a hard gelatin capsule, can be administered orally every 4-6 hours as needed. [0068] The disclosures of all cited references including publications, patents, and patent applications are expressly incorporated herein by reference in their entirety. Further, when an amount, concentration, or other value or parameter is given as either a range, preferred range, or a list of upper preferable values and lower preferable values, this is to be understood as specifically disclosing all ranges formed from any pair of any upper range limit or preferred value and any lower range limit or preferred value, regardless of whether ranges are separately disclosed. Where a range of numerical values is recited herein, unless otherwise stated, the range is intended to include the endpoints thereof, and all integers and fractions within the range. It is not intended that the scope of the invention be limited to the specific values recited when defining a range.
[0069] The present invention is further defined in the following Examples. It should be understood that these Examples, while indicating preferred embodiments of the invention, are given by way of illustration only.
Examples
Example 1
[0070] Initial testing and evaluation experiments for the immediate release ADF liquid filled capsule were based on suspending an API in PEG and filling it to weight into a hard gelatin capsule which then solidifies into a wax at room temperature. Some of these experiments used acetaminophen (APAP) as a tracer drug in place of C-II narcotics. Oxycodone HC1 and APAP are both soluble in reaerated water. The USP monograph for pooled hydrocodone bitartrate and acetaminophen tablets specifies 80% (Q) +10% release of both drugs in 30 minutes in 0.1N HC1, indicating both are capable of immediate release. As a result, APAP was expected to be a viable alternative for experimentation.
[0071 ] A formulation was prepared containing 30 mg APAP and a 50:50 ratio of PEG 3350:1500 g/mol and 0.50% FD&C dye in size 3 white opaque capsules. Three capsule fill weights were evaluated: 100 mg, 150 mg, and 200 mg. These formulations were tested for dissolution. The USP criteria for immediate release of oxycodone HC1 is 500 mL purified water as media, Q=70% at 45 minutes, Specification = 75% (Q+5%), apparatus 2 (paddles), 50 rpm. All capsule weights proved to release immediately, with the 150 mg and 200 mg formulations releasing completely at 20 minutes. 100 mg capsule fill would be preferred to decrease material costs. Table 1 below list the dissociation data of size 3 capsules containing 30 mg APAP, a 50:50 ratio of PEG 3350:1500 g/mol, and 0.5% FD&C dye.
Figure imgf000024_0001
[0072] These dosage forms contain water- and ethanol-soluble FD&C dyes, e.g., 0.5% FD&C dye, to deter extraction of the API and intravenous injection of the solution. Further rendering of the drug solution would be required to separate the pure API from the PEG and FD&C dyes.
[0073] PEG 1450 (NF grade available from Dow Chemical Company) can be used in place of PEG 1500 in the oxycodone HC1 dosage forms. Additional exemplary oxycodone HC1 dosage forms are shown in Table 2 below. 1 % citric acid may be used in the dosage forms as an API stabilizer.
Figure imgf000024_0002
Example 2 - Immediate Release ADF Liquid Fill Capsules including PEG 35000
[0074] The dissolution rate, ADF properties and melt temperatures of additional immediate release ADF oxycodone HC1 liquid fill capsule formulations containing varying amounts of PEG 1450 and PEG 35000 were evaluated. Acetaminophen (APAP) was used as a tracer drug for oxycodone HC1. The formulations are shown in Table 3 below. The target amount of APAP was 30 mg per capsule, and the target fill weight was 100 mg (batch number 92) or 200 mg (batch numbers 93-94). The capsules contained 30% w/w (batch number 92) or 15% w/w (batch numbers 93-94) APAP. Size 3 opaque hard gelatin capsules were used.
[0075] Dissolution was tested using the following criteria: Q = not less than 70% dissolved at 45 minutes, and the specification = Q+5% (75%) dissolved at 45 minutes. As shown in Table 3, the average dissolution for the three formulations ranged from 87% to 98%. Accordingly, all of the formulations met the specification of at least 75% dissolution at 45 minutes.
[0076] As mentioned above, it is generally accepted that any particle greater than 500 μιη in diameter cannot be sufficiently absorbed by the blood vessels in the nasal mucosa. Thus, in one embodiment, a formulation is considered to deter intranasal abuse if ≥ 75% of the particles are ≥ 500 μm in diameter after grinding. As shown in Table 3, the percentage of particles≥ 500 μm in diameter after grinding ranged from 90% to 92%. Thus all of the oxycodone formulations met the standard of ≥ 75% of the particles being≥ 500 μm in diameter after grinding.
Figure imgf000026_0001
[0077] The oxycodone HC1 formulations were also analyzed to determine melting temperature. The capsules were held at 40 °C / 75% relative humidity for 72 hours. As shown in Table 4 below, the batch number 92 and 93 formulations containing 100% and 82% PEG 35000, respectively, were solid at these conditions, while the batch number 94 formulation containing 59% PEG 35000 had a much softer fill.
Figure imgf000026_0002
[0078] The dissolution, particle size after grinding, and color extraction of two additional formulations of oxycodone HC1 (batch numbers 1 1 and 12) containing varying percentages of PEG 35000 and PEG 1450 were determined as described above. The formulations also contained 1 % citric acid and 14% Grey Dye. Both formulations used a size 3 opaque hard gelatin capsule with a target fill weight of 100 mg. APAP was used as a tracer drug for oxycodone HC1. The target amount of APAP was 5 mg (5% w/w) for batch number 1 1 and 30 mg (30% w/w) for batch number 12. For dissolution, Q = Not less than 70% dissolved at 45 minutes, and the specification = Q+5% (75%) dissolved at 45 minutes. As a reference, an acceptable particle size after grinding is≥ 75% particles≥ 500 μm in diameter. As a reference, an acceptable color scale designation after extraction of the dye is≥ 4 on a scale of 1 to 5, with 5 being the highest level of color.
[0079] As shown in Table 5 below, both oxycodone HC1 formulations met the criteria for dissolution rate, particle size after grinding, and color extraction.
Figure imgf000027_0001
[0080] In one embodiment, hot melt fill capsules are sufficiently viscous at elevated temperatures to allow for flow of the fill into the capsules. Accordingly, additional oxycodone HC1 formulations containing PEG 35000 and either PEG 3350 or PEG 1450 were evaluated by measuring viscosity at 75 °C at 50 rpm. Formulations were weighed out according to total wt % of a 15g batch. Each formulation was poured into a viscosity testing crucible and placed in an 80 °C water bath to melt. Once fully melted, the formulations were mixed using a stainless steel spatula and transferred to a Brookfield DV-II+ Pro Viscometer (VIS29 NCD: Upon Use) utilizing Spindle: S27 (Small Sample Adapter). The viscometer was equipped with a water jacketed crucible platform. Once the melt temperature reached 75 °C, a viscosity reading was taken in centipoise (cP). Based on manufacturer specifications, an acceptable viscosity for the purposes of this study is≤ 1000 cP. The particle size after grinding and stability at 40 °C / 75% relative humidity (RH) was also determined. For the grinding analysis, an acceptable particle size after grinding was considered to be≥ 75% particles≥ 500 μm in diameter. All
formulations were size 3 opaque hard gelatin capsules.
[0081 ] As shown in Table 6 below, batch number 100 containing 1 1 % PEG 35000 and
44% PEG 1450 had a viscosity of 1288 cP at 75 °C / 50 rpm, above the manufacturer specification of≤ 1000 cP. Accordingly, viscosity was not measured for the formulations containing higher percentages of PEG 35000 (i.e. batch numbers 97-99). In addition, batch number 100 was not sufficiently stable for storage, since this formulation was a very viscous liquid at the stability test conditions of 40 °C / 75% RH.
[0082] Although batch number 101 containing 5.5% PEG 35000 and 49.5% PEG 1450 had an acceptable viscosity (705 cP) at 75 °C / 50 rpm, the stability tests revealed that this formulation was a very viscous liquid at 40 °C / 75% RH, and thus was not stable for storage. Because the formulations containing 1 1 % PEG 35000 (batch number 100) and 5.5% PEG 35000 (batch number 101 ) were not sufficiently stable for storage, stability of batch number 103 containing 7.7% PEG 35000 was not determined.
[0083] Formulations containing PEG 35000 and PEG 3350 were also evaluated. As shown in Table 6 below, the formulation containing 6.97% PEG 35000 and 62.7% PEG 3350 (batch number 104) and the formulation containing 7.7% PEG 35000 and 69.3% PEG 3350 (batch number 105) met all of the criteria for particle size after grinding, viscosity, and stability.
Figure imgf000029_0001
Example 3 - Evaluation of Dyes [0084] Numerous dyes were evaluated for their potential to deter intravenous abuse.
Varying concentrations of FD&C Blue #2, green (FD&C Blue #2 and FD&C Yellow #5), FD&C Yellow #5, FD&C Red #40, and grey dye (FD&C Blue#l, FD&C Yellow #6, FD&C Red #40) were evaluated by dissolving them in a 95% ethanol 5% purified water (190 proof) solution and passing the solution through a syringe filter. After syringe filtering the dye solutions were visually evaluated for color intensity and rated on a scale of 0 to 5, with 0 indicating no color and 5 indicating dark, significant color. As shown in the Table 7 below, the blue and green dyes exhibited the highest color intensity at low concentrations, e.g. 0.25% w/w. Solutions of grey dye before and after filtering are shown in Figures 2A and 2B, respectively. The grey dye was particularly striking and less appealing. An acceptable color scale designation after extraction of the dye is≥ 4 on a scale of 1 to 5, with 5 being the highest level of color.
Figure imgf000030_0001
[0085] In one embodiment, the dye can be grey. Grey can be chosen because it is darker than the others and can be effective at a lower relative concentration. Grey dye can allow for the most visually deterring form with the least amount of dye present in the formulation.
Example 4 - Immediate Release ADF Oxycodone HC1 Liquid Fill Capsules
[0086] Abusers of opioid products often adulterate the product to promote more rapid release of the active ingredient. The products can be chewed and swallowed, crushed and inhaled, or extracted in water or alcohol (either crushed or intact) to produce a solution that can be used for intravenous administration or dried for insufflation of a purified product. Adulteration of the products can enable a more rapid delivery of active than can be achieved by ingestion of the intact product. This rapid onset, high exposure is associated with euphoria, drug liking, and greater abuse potential.
[0087] Current abuse-deterrent formulations have limitations. Insufflation is a common route of abuse for oxycodone HC1 products. To be attractive for insufflation, crushing a product should yield particles of less than 500 μm to allow uptake of the active substance though the nasal mucosa. Therefore, abuse deterrent formulations can be made to discourage crushing or breaking of tablets to yield particles less than 500 μm. Test methods using flat platens to crush the product as a criterion for abuse deterrence is not meaningful. All C-II narcotic drug products tested can be cut with an edged surface (e.g., scissors or a razor blade) and therefore can potentially be abused, with forces that are substantially lower than what has been reported using the breaking strength test or equivalent (e.g., >500 N). Flattening the tablets using forces greater than 500 N (with traditional "tablet breaking force" definitions) does not address abuse deterrence potential in the tested C-II narcotic drug products.
[0088] Grinding can be a better evaluation of the relative resistance of marketed products to abuse. The formulation of the present disclosure compares favorably against Roxicodone® with respect to a decrease in the percentage of particles produced after grinding that are smaller than 500 μm. Statistically different results emerge between the formulation of the present disclosure and Roxicodone® in the degree of resistance to grinding, with the formulation of the present disclosure yielding less than 50% of particles smaller than 500 μm, compared with
approximately 77% of particles less than 500 μm for Roxicodone®. Better resistance to grinding can be due to differences in the manufacturing processes and/or the excipients employed for the two products.
[0089] The formulation of the present disclosure can be resistant to abuse by nasal insufflation or extraction due to, in part, the waxy nature of the formulation contents and the solubility of the excipients. The excipients can be both water and alcohol soluble to create a formulation that makes it time consuming and costly to extract oxycodone HC1 from the formulation contents without also extracting the excipients. A high molecular weight PEG can be included because of its solubility properties (e.g., soluble in both alcohol and water) and its resistance to grinding to particle sizes of less than 500 μm. High-molecular weight PEGs are less viscous at melt temperatures than long chain PEO molecules and are soluble in both water and alcohol.
[0090] Dyes can also be used and chosen to be soluble in both water and alcohol to produce a dark colored solution upon extraction and filtering as a visual deterrent to abuse. The formulation can include the following components listed in the Table 8 below, including a number of different dyes. Table 8 below lists the components along with their solubility information taken from the various literature sources and tested experimentally (e.g., 200 proof ethanol and filtered through a 0.45 micrometer PTFE filter). The extraction of the active to a pure form can be very difficult using water or alcohol.
Figure imgf000032_0001
[0091] A conventional tablet or powder-filled capsule can be easily crushed to create a fine powder. The waxy material contained in the formulations of the present disclosure can make it difficult to manipulate into particles small enough to be easily absorbed by the nasal mucosa. The waxy material may also congeal once introduced to the semi-aqueous environment of the nasal passages, which can make it difficult to introduce the oxycodone HC1 or hydrocodone bitartrate to the bloodstream via the nasal passages.
[0092] The formulations of the present disclosure can contain one or more of the following barriers to abuse. Insufflation - The formulation can be formulated to resist grinding to particle sizes of less than about 500 μm. Extraction and Purification - The formulation can be formulated with water- and alcohol-soluble dyes to create a dark colored solution upon extraction that can be visually unappealing to intravenous drug users. The water- and alcohol- soluble excipients can present obstacles to purification of the active. In some formulations, if the solvent is flashed off or otherwise evaporated, the excipients can return to the same waxy, dark- colored form as before being introduced to the solvent. Vaporization - The formulation can contain an active, such as oxycodone HC1, which can degrade at temperatures close to where vaporization occurs. Chewing - Because the formulation is an immediate release formulation, it is not expected that crushing or cutting the dosage form will result in an especially rapid release of the drug to produce a "euphoric high."
[0093] Table 9 below lists exemplary formulations for the oxycodone HC1 abuse deterrent formulation capsules.
Figure imgf000034_0001
[0094] Formulations of the present disclosure were manufactured by the following exemplary process. The components of the hot-melt suspension, consisting of Polyethylene Glycol 3350, Polyethylene Glycol 35000, Dye Blend, Grey Powder, Citric Acid and Oxycodone HC1 were dispensed according to theoretical batch quantities based on formulation weight percents.
[0095] Polyethylene Glycol 3350, Polyethylene Glycol 35000, Dye Blend, Grey Powder, Oxycodone HC1 and Citric Acid were added to an Olsa 150 Liter Kettle and heated to a temperature of 70 ± 20 °C. Utilizing the homogenizer mixer, external anchor blades and internal mixing blades, the melt was then mixed until uniform
[0096] Prior to transferring the hot-melt suspension from the kettle to the Shionogi F40 capsule filling machine hopper, a transfer pump and three heat traced hoses were set up and the melt/suspension was recirculated. Mixing and recirculating continued until capsule filling was completed. [0097] The Shionogi F40 capsule filling machine target fill weight was set with an Action Limit of ± 3.5% and a Control Limit of ± 5.0% plus the average empty capsule weight.
[0098] In-process capsule samples were taken at the beginning, end, and every 30 minutes (for the average capsule weight of 15 filled capsules). Filled capsules were placed onto stainless steel cooling trays and allowed to cure. Following curing, 100% capsule weight inspection was performed using a Shionogi capsule weight inspection machine An exemplary manufacturing process is shown in Figure 3.
[0099] The formulations of the present disclosure are stable upon storage at 25, 30, 35, 40 or 45 °C, and at 60%, 65%, 70% or 75% relative humidity, e.g., 30 °C / 65% RH or 40 °C / 75% RH. The formulation of the present disclosure can be stable under any of these conditions for up to 1 , 2, 3, 4, 5, 6, 9, 12, 16, 18, 24, or 36 months.
Example 5 - Abuse Deterrent Properties of Immediate Release Liquid Fill Capsule PEG Formulations
[00100] In one embodiment, there are at least three determining factors which deem an immediate release drug product "abuse deterrent," namely resistance to grinding, purity upon extraction, and visual evaluation following extraction. Cutting the dosage form can be performed in order to increase the surface area of the product prior to ingesting it in an effort to increase the rate of dissolution into the digestive tract. Cutting can also be used to increase the efficiency of grinding or extraction. Cutting alone, however, is not sufficient to render a formulation abusable. Grinding the dosage form can be performed in order to decrease the particle size of the product more efficiently than cutting in an effort to insufflate (snort) for immediate release into the blood vessels of the nasal passages. A readily available tool used for grinding is a commercially available coffee grinder. In one embodiment, a drug product is considered abuse deterrent if the % material in the pan (≤500μm) is≤ 50%. A dosage form which, when ground, produces≤ 50% of the material on a per-dosage form basis available for nasal insufflation (≤ 500 μm) is considered abuse deterrent. The purpose of this study was to determine the grinding potential of different dosage forms of oxycodone HC1. Texture analysis is the mechanical testing of pharmaceutical products in order to measure their physical properties. The Retsch Knife Mill GRINDOMIX GM200 (TE96) was utilized to mimic a commercially available coffee grinder (Mr. Coffee) in order to grind the drug products into a particle size that is suitable for intranasal abuse (insufflation). Particle size analysis was conducted utilizing an ATM L3P Sonic Sifter (TE47), utilizing a 500 micrometer (μm) particle size sieve (35 mesh). For the purposes of this study, any particle less than 500 μm in diameter is considered suitable for intranasal abuse. It is generally accepted that any particle greater than 500 μm in diameter cannot be sufficiently absorbed by the blood vessels in the nasal passages.
[00101] The Retsch Knife Mill GRINDOMIX GM200 utilizes a circular blade attachment to mimic commercially available coffee grinders. The GM200 has a top speed of 10,000 revolutions per minute (rpm), while commercially available coffee grinders have a top speed of approximately 20,000 rpm (an approximate two-fold increase in speed when comparing the GM200 to a Mr. Coffee grinder). However, the approximate two-fold increase in blade diameter (1 18 mm vs. 60 mm, when comparing the GM200 to a Mr. Coffee grinder, respectively) compensates for the approximate twofold decrease in top speed via the inversely proportional relationship of the two variables. Further, the torque provided by the GM200 is significantly higher than the torque provided by a Mr. Coffee grinder (0.860 Nm (Newton meters) of the GM200 vs. 0.062 Nm of the Mr. Coffee grinder, respectively), which additionally illustrates the ability (or lack thereof) of the Mr. Coffee grinder to modify the drug products into a particle size suitable for intranasal abuse. The study evaluated the difference in particle sizes of several different formulations of oxycodone HC1 following modification (grinding) by the GM200.
[00102] Experimental: The samples tested are formulated according to Table 9. The following test equipment was used: Retsch Knife Mill GRINDOMIX GM200 (TE96), ATM L3P Sonic Sifter (TE47), and a 500 μm sieve (35 mesh). The following testing conditions were used: Analysis speed: 10,000 rpm; Analysis time: 30 seconds; Sieve Size: 500 μm (35 mesh); Analysis time: 1 minutes (no pulse). Each sample was prepared in triplicate (N=3).
[00103] The composite sample was transferred to a tared weigh boat and the weight of the sample was recorded. The following equation was used to calculate the % sample loss:
Figure imgf000036_0001
[00104] The weight of the 35 mesh sieve and sample pan was recorded. The testing apparatus was assembled with the 35 mesh sieve above the sample pan. The composite sample was transferred to the testing apparatus and analyzed utilizing the following parameters: 1 minute analysis time and no pulse. The analyzed 35 mesh sieve and sample pan were weighed. The % material remaining on the 35 mesh sieve (≥ 500 μm) and in the sample pan (≤ 500 μm) was calculated using the following equation: x 100
Figure imgf000037_0001
[00105] Table 10 below shows the particle size after grinding for the oxycodone HC1 formulations tested. During testing it was observed by visual observation that the capsule portion of the dosage form of all evaluated batches was not being significantly modified by TE96, and that the majority of the capsule portion remained in the 35 mesh sieve (≥ 500μm). The grinding / particle size analysis for this protocol is based on weight differences, which, when the capsule portion is taken into account, can skew the results towards a higher proportion of particles≥ 500 μm.
[00106] In order to confirm the particle size of capsules modified by TE96, three empty size 3 capsules (N=1) were ground and analyzed. Table 1 1 shows that for size 3 capsules, 99% of the particles by weight were≥ 500 μm. Additional calculations were made which compensated for the percentage of capsules≥ or≤ 500 μm. These calculations removed the average capsule weight from the analyzed sample by subtracting it from the we<ght≥ 500 μm and the weight ≤500 μm. The results adjusted for capsule weight are shown in Table 12.
Figure imgf000038_0001
Figure imgf000038_0002
[00107] As shown in Table 12, after adjusting for the capsule portion, the average percentage of particles≥ 500 μm after grinding for the oxycodone HC1 capsules ranged from 62.3% to 68.2%. For comparison, approximately 20% of particles by weight of an Immediate Release (IR) Roxicodone® formulation were≥ 500 μm after the same grinding procedure.
Figure imgf000039_0001
[00108] Table 13 summarizes the grinding results and statistical analysis of the % material≤ 500 μm for the Present Disclosure 15mg and Roxicodone® 15mg tablets (Mallinckrodt
Pharmaceuticals, Inc.).
Figure imgf000040_0001
[00109] Another method of rending a drug product abusable is via extraction of the active substance from the dosage form to produce a pure residue. This method can be performed, and is often performed, using a high proof alcohol or an aqueous media. The formulation of the present disclosure can be readily soluble in both aqueous and alcohol environments when the contents are removed from the capsule. Therefore, aqueous and alcohol extraction techniques were evaluated. Solutions were analyzed qualitatively for solution color following filtration, as well as quantitatively for % label claim (LC) (with regards to oxycodone HCI) of solution following filtration. Additionally, evaporated residual samples were analyzed qualitatively for residue color following evaporation, as well as quantitatively for purity determination following the %LC calculations. The quantitative results of the analysis determine the % purity (with regards to oxycodone HCI) of the extracted sample solution described above. A drug product can be considered abuse deterrent if the % residue purity is≤ 50%. In other embodiments, less than or equal to 40%, 45%, 55%, 60%, 65%, 70% or 75%. Residue purity levels (with regards to the API)≤ 50% can infer that the excipient load is greater than the API level contained in the residue. In one embodiment, this can be considered abuse deterrent with regards to potential intravenous abuse of a purified residue. Using the data analysis software functionality of Microsoft Excel and a 95% significance interval (p-value = 0.05), the F-test and t-tests was analyzed in order to determine if the drug products provide statistically different % purity values.
[001 10] Tables 14 and 15 show the formulation of the present disclosure results in 9% and 9% purity with regards to oxycodone HC1, in alcohol and aqueous environments, respectively. This is in comparison to Roxicodone® 15mg, which has a purity of 68% and 19% purity in alcohol and aqueous environments, respectively. This data proves the formulation of the present disclosure is statistically different than Roxicodone18 in both alcohol and aqueous extracts.
Figure imgf000041_0001
[001 1 1 ] Color is one identifying characteristic of commercial drug products. Color can be applied to the dosage form in two ways: dye or coating. High potency alcohol (i.e.,≥190 proof (95%)) is one extraction solvent that can be used by abusers for APIs which are insoluble in water or in order to separate the API from other water soluble excipients. Dyes or coatings can potentially be used to alter the physical appearance of the extracted solution of drug product (i.e., turn the resulting solution a noticeable color).
[001 12] Accordingly, the inclusion of one or more dyes in a drug formulation is one method to render a formulation abuse deterrent. Significant discoloration of an extraction product from a formulation subject to abuse can discourage a potential abuser from using (e.g., injecting or ingesting) the extraction product.
[001 13] A study was conducted to investigate the effect of dyes in the formulations of the present disclosure. Extraction products from whole formulations were visually inspected to determine abuse deterrence following alcohol extraction. Capsules were added to a flask containing 190 proof ethanol and shaken at 250 rpm for 3 hours. After 3 hours all capsule contents were fully dissolved. Solutions were filtered with a syringe filter and then visually analyzed for color intensity. The samples tested were the immediate release oxycodone HC1 capsules according to Table 9 above.
[001 14] The unfiltered and filtered solutions are shown in Figures 4 and 5, respectively. As shown in Table 16 below, all of the filtered solutions had a color value of 5, indicating that all seven evaluated batches produced a filtered solution which was significantly dark in color. This significant dark color provides potential abuse deterrence to CII narcotic drug products.
Figure imgf000042_0001
[001 15] Additionally, the color of filtered solutions and resulting evaporated residues of alcoholic and aqueous extracts of the formulations of the present disclosure and Roxicodone® were compared. Table 17 below shows both of these dosage forms, with the formulation of the present disclosure providing the most visual deterrence for both the filtered solution and evaporated residue in both media. Table 1 7
Figure imgf000043_0001
[001 16] While this disclosure has been particularly shown and described with reference to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims

We claim:
1. An immediate release, abuse deterrent capsule comprising:
(a) an active substance susceptible to abuse;
(b) a first polyethylene glycol (PEG) having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons; and
(c) a second PEG having an average molecular weight between about 3000 Daltons and about 4000 Daltons, wherein the ratio of the first PEG to the second PEG is less than about 1 :4 w/w.
2. The capsule of claim 1 , wherein the first PEG and the second PEG together are at least about 60 wt% of the dosage form.
3. The capsule of claim 1, wherein at least 80% of the contents are soluble both water and
alcohol.
4. The capsule of claim 1, wherein the active substance is hydrocodone bitartrate.
5. The capsule of claim 1, wherein the active substance is oxycodone HC1.
6. The capsule of claim 1 , further comprising a grey dye comprising FD&C Blue #1, FD&C Yellow #6, and FD&C Red #40.
7. The capsule of claim 6, wherein the dye provides a visual deterrent to abuse.
8. The capsule of claim 1, wherein the ratio of the first PEG to the second PEG is between
about 1 :7 w/w and about 1 :1 1 w/w.
9. The capsule of claim 1, wherein the first PEG has an average molecular weight of about 35,000 Daltons and the second PEG has an average molecular weight of about 3350 Daltons.
10. The capsule of claim 1, wherein the capsule comprises at least about 2.5 wt% of the active substance.
1 1. The capsule of claim 1, wherein the capsule is prepared by filling a capsule body with a
heated homogenized suspension comprising the active substance, the first PEG and the second PEG.
12. An immediate release, abuse deterrent capsule comprising:
(a) an active substance susceptible to abuse; and
(b) PEG with a weighted average molecular weight between about 6200 Daltons and about 7800 Daltons.
13. The capsule of claim 12, wherein the capsule comprises at least about 60 wt% of the PEG.
14. The capsule of claim 12, wherein the active substance is hydrocodone bitartrate.
15. The capsule of claim 12, wherein the active substance is oxycodone HC1.
16. An immediate release, abuse deterrent capsule comprising:
(a) an active substance susceptible to abuse;
(b) a first PEG having a melting point greater than or equal to about 60 °C ; and
(c) a second PEG having a melting point less than or equal to about 57 °C;
wherein the contents of the capsule are solid at 40 °C / 75% relative humidity, and at least 75% of the active ingredient is released from the capsule within 45 minutes following administration or dissolution testing.
17. The capsule of claim 16, wherein the first PEG and the second PEG together are at least about 60 wt% of the capsule.
18. The capsule of claim 16, wherein the active substance is hydrocodone bitartrate.
19. The capsule of claim 16, wherein the active substance is oxycodone HC1.
20. A process for the production of an immediate release, abuse deterrent capsule comprising at least one active substance susceptible to abuse comprising:
(a) preparing a homogenized suspension of:
(i) the at least one active substance susceptible to abuse;
(ii) a first PEG having an average molecular weight between about 30,000 Daltons and about 40,000 Daltons; and
(iii) a second PEG having an average molecular weight between about 3000
Daltons and about 4000 Daltons; and (b) filling the homogenized suspension into a capsule body to produce the capsule, wherein the ratio of the first PEG to the second PEG is less than about 1 :4 w/w.
21. The process of claim 20, wherein the first PEG and the second PEG together are at least about 60 wt% of the capsule.
22. The process of claim 20, wherein the active substance is hydrocodone bitartrate.
23. The process of claim 20, wherein the active substance is oxycodone HC1.
24. The process of claim 20, wherein the capsule is formed by joining a capsule body with a capsule cap.
25. A method of treating pain comprising administering to a subject in need thereof a
therapeutically effective amount of the capsule of claim 1.
PCT/US2015/039336 2014-07-17 2015-07-07 Immediate release abuse deterrent liquid fill dosage form WO2016010771A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
ES15821947T ES2809458T3 (en) 2014-07-17 2015-07-07 Liquid filled, abuse deterrent and immediate release dosage form
CA2955229A CA2955229C (en) 2014-07-17 2015-07-07 Immediate release abuse deterrent liquid fill dosage form
AU2015290098A AU2015290098B2 (en) 2014-07-17 2015-07-07 Immediate release abuse deterrent liquid fill dosage form
JP2017502652A JP6371463B2 (en) 2014-07-17 2015-07-07 Immediate release abuse deterrent liquid filler form
EP15821947.7A EP3169315B1 (en) 2014-07-17 2015-07-07 Immediate release abuse deterrent liquid fill dosage form
DK15821947.7T DK3169315T3 (en) 2014-07-17 2015-07-07 Liquid-filled dosage form to prevent immediate release abuse
AU2019200026A AU2019200026A1 (en) 2014-07-17 2019-01-03 Immediate release abuse deterrent liquid fill dosage form

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462025878P 2014-07-17 2014-07-17
US62/025,878 2014-07-17

Publications (1)

Publication Number Publication Date
WO2016010771A1 true WO2016010771A1 (en) 2016-01-21

Family

ID=55073631

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/039336 WO2016010771A1 (en) 2014-07-17 2015-07-07 Immediate release abuse deterrent liquid fill dosage form

Country Status (8)

Country Link
US (2) US9707184B2 (en)
EP (1) EP3169315B1 (en)
JP (2) JP6371463B2 (en)
AU (2) AU2015290098B2 (en)
CA (1) CA2955229C (en)
DK (1) DK3169315T3 (en)
ES (1) ES2809458T3 (en)
WO (1) WO2016010771A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3209282A4 (en) * 2014-10-20 2018-05-23 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4450877A (en) * 1977-11-03 1984-05-29 Hoechst Aktiengesellschaft Pharmaceutical preparations in solid unit dosage form
WO1991007950A1 (en) * 1989-11-24 1991-06-13 Farmitalia Carlo Erba Limited Pharmaceutical compositions
WO2008021394A2 (en) * 2006-08-15 2008-02-21 Theraquest Biosciences, Llc Pharmaceutical formulations of cannabinoids and method of use
US20090123386A1 (en) 2005-04-06 2009-05-14 Victor Morrison Young Abuse Resistant Capsules
US20100204259A1 (en) 2009-02-06 2010-08-12 Egalet A/S Immediate release composition resistant to abuse by intake of alcohol
US20140010873A1 (en) 2012-07-06 2014-01-09 Egalet Ltd. Abuse deterrent pharmaceutical compositions for controlled release
US20140171481A1 (en) * 2010-06-10 2014-06-19 Abbvie, Inc. Solid compositions

Family Cites Families (689)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA264736A (en) 1926-10-05 S. Goulet William Rotary engine
CA265145A (en) 1926-10-19 T. Thomas Horace Transmission brake
CA265559A (en) 1926-11-02 F. L. Smidth And Co. Tube mill
GB135381A (en)
NL143131B (en) 1964-07-11 1974-09-16 Acf Chemiefarma Nv PROCEDURE FOR THE PREPARATION OF ANTIPARASITARY ACTIVE PREPARATIONS AND OF ACTIVE INGREDIENTS THEREOF, AND THE PREPARATIONS THEREFORE OBTAINED.
CS162957B1 (en) 1972-05-24 1975-07-31
US4126684A (en) 1976-02-11 1978-11-21 Ciba-Geigy Corporation 4-amino-3-p-halophenylbutyric acids and their derivatives used in the control of narcotic abuse
JPS5584166A (en) 1978-12-20 1980-06-25 Lion Hamigaki Kk Band for spongy medicine
US4507276A (en) 1982-08-20 1985-03-26 Bristol-Myers Company Analgesic capsule
PL133984B2 (en) 1983-02-21 1985-07-31 Method of manufacture of complex of derivatives of cellulose with polyhydric alcohol or its polymer
JPS6092214A (en) 1983-10-26 1985-05-23 Ss Pharmaceut Co Ltd Composition for filling soft capsule
JPH0713012B2 (en) 1984-02-08 1995-02-15 ア−ル ピ− シ−ラ− コ−ポレ−シヨン Acetaminophen / gelatin capsule
US6309669B1 (en) 1984-03-16 2001-10-30 The United States Of America As Represented By The Secretary Of The Army Therapeutic treatment and prevention of infections with a bioactive materials encapsulated within a biodegradable-biocompatible polymeric matrix
US5155212A (en) 1986-05-21 1992-10-13 Abbott Laboratories Phencyclidine and phencyclidine metabolites assay, tracers, immunogens, antibodies and reagent kit
US5071643A (en) * 1986-10-17 1991-12-10 R. P. Scherer Corporation Solvent system enhancing the solubility of pharmaceuticals for encapsulation
US5202128A (en) 1989-01-06 1993-04-13 F. H. Faulding & Co. Limited Sustained release pharmaceutical composition
US5141961A (en) 1991-06-27 1992-08-25 Richrdson-Vicks Inc. Process for solubilizing difficulty soluble pharmaceutical actives
US5266331A (en) 1991-11-27 1993-11-30 Euroceltique, S.A. Controlled release oxycodone compositions
US5681585A (en) 1991-12-24 1997-10-28 Euro-Celtique, S.A. Stabilized controlled release substrate having a coating derived from an aqueous dispersion of hydrophobic polymer
US5518730A (en) 1992-06-03 1996-05-21 Fuisz Technologies Ltd. Biodegradable controlled release flash flow melt-spun delivery system
NZ256346A (en) 1992-10-09 1997-04-24 Procter & Gamble Medicaments containing 3-l-menthoxypropane-1,2-diol for treating cold symptoms
WO1994018970A1 (en) 1993-02-18 1994-09-01 Warner-Lambert Company Cold/sinus preparation consisting of phenindamine tartarate with or without therapeutic agents
IL108366A (en) 1993-03-11 1999-11-30 Taro Vit Ind Ltd Pharmaceutical compositions in semisolid form and a squeezable container for administration thereof
SE9301057L (en) 1993-03-30 1994-10-01 Pharmacia Ab Controlled release preparation
US5431916A (en) 1993-04-29 1995-07-11 The Procter & Gamble Company Pharmaceutical compositions and process of manufacture thereof
CA2161217A1 (en) 1993-04-30 1994-11-10 Carmelita Macklin Russell Coated pharmaceutical compositions
WO1994026731A1 (en) 1993-05-13 1994-11-24 Merck Frosst Canada Inc. 2-substituted-3,4-diarylthiophene derivatives as inhibitors of cyclooxygenase
JPH1133084A (en) 1994-02-10 1999-02-09 Yamanouchi Pharmaceut Co Ltd Intraoral soluble type tablet and manufacture thereof
US5458879A (en) 1994-03-03 1995-10-17 The Procter & Gamble Company Oral vehicle compositions
GB9422154D0 (en) 1994-11-03 1994-12-21 Euro Celtique Sa Pharmaceutical compositions and method of producing the same
US5965161A (en) 1994-11-04 1999-10-12 Euro-Celtique, S.A. Extruded multi-particulates
US5660859A (en) 1994-12-29 1997-08-26 Mcneil-Ppc, Inc. Gelling agent for polyethylene glycol
US5616621A (en) 1995-01-30 1997-04-01 American Home Products Corporation Taste masking liquids
US5840731A (en) 1995-08-02 1998-11-24 Virginia Commonwealth University Pain-alleviating drug composition and method for alleviating pain
WO1997020561A1 (en) 1995-12-07 1997-06-12 Eli Lilly And Company Composition for treating pain
CN1208348A (en) 1995-12-07 1999-02-17 伊莱利利公司 Method for treating pain
US6159501A (en) 1996-03-08 2000-12-12 Nycomed Danmark A/S Modified release multiple-units dosage composition for release of opioid compounds
WO1997033566A2 (en) 1996-03-12 1997-09-18 Alza Corporation Composition and dosage form comprising opioid antagonist
WO1997045091A2 (en) 1996-05-31 1997-12-04 Euro-Celtique, S.A. Sustained release oxycodone formulations with no fed/fast effect
AU3508097A (en) 1996-06-26 1998-01-14 Board Of Regents, The University Of Texas System Hot-melt extrudable pharmaceutical formulation
US6024980A (en) 1996-06-28 2000-02-15 Mcneil-Ppc, Inc. Multiphase soft gelatin dosage form
US5914129A (en) 1996-07-23 1999-06-22 Mauskop; Alexander Analgesic composition for treatment of migraine headaches
US6190591B1 (en) 1996-10-28 2001-02-20 General Mills, Inc. Embedding and encapsulation of controlled release particles
DE19651551C2 (en) 1996-12-11 2000-02-03 Klinge Co Chem Pharm Fab Opioid antagonist-containing galenic formulation
US6160020A (en) 1996-12-20 2000-12-12 Mcneill-Ppc, Inc. Alkali metal and alkaline-earth metal salts of acetaminophen
HUP9903375A3 (en) 1997-03-24 2000-04-28 Lilly Co Eli Synergistic pharmaceutical compositions containing olanzapine and analgetic drugs
CA2289190A1 (en) 1997-05-07 1998-11-12 Algos Pharmaceutical Corporation Composition and method combining an antidepressant with an nmda receptor antagonist, for treating neuropathic pain
WO1998050075A1 (en) 1997-05-07 1998-11-12 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with nmda-blockers for treating pain
US20040043071A1 (en) 2002-06-21 2004-03-04 Pauletti Giovanni M. Intravaginal mucosal or transmucosal delivery of antimigraine and antinausea drugs
US7744916B2 (en) 1997-06-11 2010-06-29 Umd, Inc. Coated vaginal device for delivery of anti-migraine and anti-nausea drugs
WO1999007413A1 (en) 1997-08-11 1999-02-18 Algos Pharmaceutical Corporation Substance p inhibitors in combination with nmda-blockers for treating pain
RS49982B (en) 1997-09-17 2008-09-29 Euro-Celtique S.A., Synergistic analgesic combination of opioid analgesic and cyclooxygenase-2 inhibitor
US20030185761A1 (en) 1997-10-01 2003-10-02 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating pain
CN1204890C (en) 1997-12-22 2005-06-08 欧罗赛铁克股份有限公司 Method for preventing abuse of opioid dosage forms
RU2241458C2 (en) 1997-12-22 2004-12-10 Эро-Селтик, С.А. Combinations of agonist/antagonist for opioid
US6251430B1 (en) 1998-02-04 2001-06-26 Guohua Zhang Water insoluble polymer based sustained release formulation
AU2587299A (en) 1998-02-06 1999-08-23 Union Carbide Chemicals & Plastics Technology Corporation Alkylene oxide polymer compositions
US6245357B1 (en) 1998-03-06 2001-06-12 Alza Corporation Extended release dosage form
AU747549B2 (en) 1998-03-23 2002-05-16 General Mills Inc. Encapsulation of components into edible products
US6054451A (en) 1998-04-21 2000-04-25 Algos Pharmaceutical Corporation Analgesic composition and method for alleviating pain
AU777977B2 (en) 1998-10-09 2004-11-04 General Mills Inc. Encapsulation of sensitive liquid components into a matrix to obtain discrete shelf-stable particles
US20060240105A1 (en) 1998-11-02 2006-10-26 Elan Corporation, Plc Multiparticulate modified release composition
AU1398899A (en) 1998-11-12 2000-06-05 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with centrally acting analgesics
CA2351224A1 (en) 1998-11-12 2000-05-25 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with nmda-blockers for treating pain
FR2787715B1 (en) 1998-12-23 2002-05-10 Synthelabo PHARMACEUTICAL COMPOSITION COMPRISING A HYPNOTIC COMPOUND OR ONE OF ITS PHARMACEUTICALLY ACCEPTABLE SALTS
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6716452B1 (en) 2000-08-22 2004-04-06 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US7060708B2 (en) 1999-03-10 2006-06-13 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
IT1311924B1 (en) 1999-04-13 2002-03-20 Nicox Sa PHARMACEUTICAL COMPOUNDS.
IT1311923B1 (en) 1999-04-13 2002-03-20 Nicox Sa PHARMACEUTICAL COMPOUNDS.
US6500459B1 (en) 1999-07-21 2002-12-31 Harinderpal Chhabra Controlled onset and sustained release dosage forms and the preparation thereof
AU6381300A (en) 1999-07-29 2001-02-19 Roxane Laboratories, Inc. Opioid sustained-released formulation
IT1313579B1 (en) 1999-07-30 2002-09-09 Acraf PARACETAMOL-BASED LIQUID PHARMACEUTICAL COMPOSITION.
IT1314184B1 (en) 1999-08-12 2002-12-06 Nicox Sa PHARMACEUTICAL COMPOSITIONS FOR THE THERAPY OF STRESS-OXIDATIVE CONDITIONS
CA2388560A1 (en) 1999-08-31 2001-03-08 Grunenthal Gmbh Sustained-release form of administration containing tramadol saccharinate
US6432450B1 (en) 1999-09-09 2002-08-13 Gerhard Gergely Effervescent granules with delayed effervescent effect
US6264983B1 (en) 1999-09-16 2001-07-24 Rhodia, Inc. Directly compressible, ultra fine acetaminophen compositions and process for producing same
US6572891B1 (en) 1999-10-23 2003-06-03 Alkaloid Ad Sublingual oral dosage form
EP1231869B1 (en) 1999-10-29 2005-09-21 Children's Hospital of Los Angeles Bone hemostasis method and materials
WO2001032928A2 (en) 1999-11-05 2001-05-10 Phase-1 Molecular Toxicology Methods of determining individual hypersensitivity to an agent
US20050233459A1 (en) 2003-11-26 2005-10-20 Melker Richard J Marker detection method and apparatus to monitor drug compliance
US6207674B1 (en) 1999-12-22 2001-03-27 Richard A. Smith Dextromethorphan and oxidase inhibitor for weaning patients from narcotics and anti-depressants
US20020055512A1 (en) 2000-01-21 2002-05-09 Cortendo Ab. Compositions for delivery of a cortisol antagonist
US6608041B2 (en) 2000-02-18 2003-08-19 Milton Hammerly Analgesics combined with naturally-occurring chondroprotective agents
US20010036943A1 (en) 2000-04-07 2001-11-01 Coe Jotham W. Pharmaceutical composition for treatment of acute, chronic pain and/or neuropathic pain and migraines
US6955821B2 (en) 2000-04-28 2005-10-18 Adams Laboratories, Inc. Sustained release formulations of guaifenesin and additional drug ingredients
JP2004515455A (en) 2000-05-05 2004-05-27 ペイン・セラピューティクス・インコーポレイテッド Opioid antagonist compositions and dosage forms
WO2001085150A2 (en) 2000-05-05 2001-11-15 Pain Therapeutics, Inc. Opioid antagonist containing composition for enchaching the potency or reducing adverse side effects ofopioid agonists
US20040156872A1 (en) 2000-05-18 2004-08-12 Elan Pharma International Ltd. Novel nimesulide compositions
AU2001264570A1 (en) 2000-05-31 2001-12-11 Warner Lambert Company Combinations of an endothelin receptor antagonist and an antiepileptic compound having pain alleviating properties or analgesic
DE10029201A1 (en) 2000-06-19 2001-12-20 Basf Ag Retarded release oral dosage form, obtained by granulating mixture containing active agent and polyvinyl acetate-polyvinyl pyrrolidone mixture below the melting temperature
GB0015617D0 (en) 2000-06-26 2000-08-16 Vectura Ltd Improved preparations for dermal delivery of active substances
US20040022787A1 (en) 2000-07-03 2004-02-05 Robert Cohen Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US6740333B2 (en) 2000-07-07 2004-05-25 Anestic Aps Suppository and composition comprising at least one polyethylene glycol
CA2386794A1 (en) 2000-07-13 2002-01-24 Euro-Celtique, S.A. Salts and bases of 17-(cyclopropylmethyl)-4,5 alpha-epoxy-6-methylenemorphinan-3,14 diol for optimizing dopamine homeostasis during administration of opioid analgesics
WO2003020200A2 (en) 2000-11-16 2003-03-13 New River Pharmaceuticals Inc. A novel pharmaceutical compound and methods of making and using same
WO2002051432A1 (en) 2001-11-16 2002-07-04 New River Pharmaceuticals Inc. A novel pharmaceutical compound and methods of making and using same
WO2003079972A2 (en) 2002-02-22 2003-10-02 New River Parmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US20020099013A1 (en) 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
US6924273B2 (en) 2000-10-03 2005-08-02 Scott W. Pierce Chondroprotective/restorative compositions and methods of use thereof
IL139177A0 (en) 2000-10-20 2001-11-25 S C Republic Dev S R L Sustained release drug delivery system
CN101317825A (en) 2000-10-30 2008-12-10 欧罗赛铁克股份有限公司 Controlled release hydrocodone formulations
US8394813B2 (en) 2000-11-14 2013-03-12 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
US6800668B1 (en) 2001-01-19 2004-10-05 Intellipharmaceutics Corp. Syntactic deformable foam compositions and methods for making
US6869618B2 (en) 2001-04-10 2005-03-22 Kiel Laboratories, Inc. Process for preparing tannate liquid and semi-solid dosage forms
UA81224C2 (en) 2001-05-02 2007-12-25 Euro Celtic S A Dosage form of oxycodone and use thereof
US20110104214A1 (en) 2004-04-15 2011-05-05 Purdue Pharma L.P. Once-a-day oxycodone formulations
US20030004177A1 (en) 2001-05-11 2003-01-02 Endo Pharmaceuticals, Inc. Abuse-resistant opioid dosage form
JP2004534056A (en) 2001-06-08 2004-11-11 エンドー ファーマシューティカルズ, インコーポレイティド Controlled release dosage forms using acrylic polymers and processes for making the same
EP1399151A4 (en) 2001-06-11 2004-08-04 Merck & Co Inc A method for treating inflammatory diseases by administering a ppar-delta agonist
US7968119B2 (en) 2001-06-26 2011-06-28 Farrell John J Tamper-proof narcotic delivery system
WO2003004009A1 (en) 2001-07-02 2003-01-16 Geneva Pharmaceuticals, Inc. Pharmaceutical composition
SI1416842T1 (en) 2001-07-18 2009-06-30 Euro Celtique Sa Pharmaceutical combinations of oxycodone and naloxone
WO2003013481A1 (en) 2001-08-03 2003-02-20 Bakulesh Mafatlal Khamar The process of manufacturing pharmaceutical composition with increased content of poorly soluble pharmaceutical ingredients
US20030068375A1 (en) 2001-08-06 2003-04-10 Curtis Wright Pharmaceutical formulation containing gelling agent
US7141250B2 (en) 2001-08-06 2006-11-28 Euro-Celtique S.A. Pharmaceutical formulation containing bittering agent
WO2003015531A2 (en) 2001-08-06 2003-02-27 Thomas Gruber Pharmaceutical formulation containing dye
US7842307B2 (en) 2001-08-06 2010-11-30 Purdue Pharma L.P. Pharmaceutical formulation containing opioid agonist, opioid antagonist and gelling agent
US20030044458A1 (en) 2001-08-06 2003-03-06 Curtis Wright Oral dosage form comprising a therapeutic agent and an adverse-effect agent
US7375083B2 (en) 2003-09-30 2008-05-20 Shire Llc Pharmaceutical compositions for prevention of overdose or abuse
US20030068276A1 (en) 2001-09-17 2003-04-10 Lyn Hughes Dosage forms
US20030092724A1 (en) 2001-09-18 2003-05-15 Huaihung Kao Combination sustained release-immediate release oral dosage forms with an opioid analgesic and a non-opioid analgesic
US20040234602A1 (en) 2001-09-21 2004-11-25 Gina Fischer Polymer release system
US20040253310A1 (en) 2001-09-21 2004-12-16 Gina Fischer Morphine polymer release system
CA2459976A1 (en) 2001-09-26 2003-04-03 Penwest Pharmaceuticals Company Opioid formulations having reduced potential for abuse
US8603514B2 (en) 2002-04-11 2013-12-10 Monosol Rx, Llc Uniform films for rapid dissolve dosage form incorporating taste-masking compositions
US7666337B2 (en) 2002-04-11 2010-02-23 Monosol Rx, Llc Polyethylene oxide-based films and drug delivery systems made therefrom
US7056500B2 (en) 2001-10-18 2006-06-06 Nektar Therapeutics Al, Corporation Polymer conjugates of opioid antagonists
WO2003034991A2 (en) 2001-10-22 2003-05-01 Taro Pharmaceutical Industries Ltd. Taste masking spill-resistant formulation
US20030235618A1 (en) 2001-10-22 2003-12-25 Taro Pharmaceutical Industries Ltd. Taste masking spill-resistant formulation
US20050143471A1 (en) 2001-10-22 2005-06-30 Shen Gao Taste masking spill-resistant formulation
US6771370B2 (en) 2001-10-22 2004-08-03 The Texas A&M University System Characterizing powders using frequency-domain photon migration
CA2464528A1 (en) 2001-11-02 2003-05-15 Elan Corporation, Plc Pharmaceutical composition
US8329217B2 (en) 2001-11-06 2012-12-11 Osmotica Kereskedelmi Es Szolgaltato Kft Dual controlled release dosage form
US20050154046A1 (en) 2004-01-12 2005-07-14 Longgui Wang Methods of treating an inflammatory-related disease
WO2003051878A1 (en) 2001-12-14 2003-06-26 Merck Frosst Canada & Co. Quinolinones as prostaglandin receptor ligands
US6713470B2 (en) 2002-01-22 2004-03-30 Ml Laboratories Plc Method of treatment
US20070167853A1 (en) 2002-01-22 2007-07-19 Melker Richard J System and method for monitoring health using exhaled breath
KR100540035B1 (en) 2002-02-01 2005-12-29 주식회사 태평양 Multi-stage oral drug controlled-release system
US20040029864A1 (en) 2002-02-04 2004-02-12 Pharmacia Corporation Treatment of colds and cough with a combination of a cyclooxygenase-2 selective inhibitor and a colds and cough active ingredient and compositions thereof
ES2271542T3 (en) 2002-02-07 2007-04-16 Pharmacia Corporation PHARMACEUTICAL DOSAGE FORM FOR ADMINISTRATION BY THE MUCOSA.
JP4174426B2 (en) 2002-02-07 2008-10-29 ファルマシア コーポレーション Pharmaceutical tablets
GB0203276D0 (en) 2002-02-12 2002-03-27 Novartis Ag Organic compounds
US20060177381A1 (en) 2002-02-15 2006-08-10 Howard Brooks-Korn Opiopathies
CA2476201C (en) 2002-02-21 2009-09-01 Biovail Laboratories Incorporated Modified release formulations of at least one form of tramadol
US6753009B2 (en) 2002-03-13 2004-06-22 Mcneil-Ppc, Inc. Soft tablet containing high molecular weight polyethylene oxide
EP1578350B1 (en) 2002-03-26 2009-05-27 Euro-Celtique S.A. Sustained-release gel coated compositions
DE10215131A1 (en) 2002-04-05 2003-10-16 Euro Celtique Sa Stable pharmaceutical preparation useful for the treatment of pain, especially severe pain, without causing side-effects comprises oxycodone and naxolone in retarded release formulation
DE10215067A1 (en) 2002-04-05 2003-10-16 Euro Celtique Sa Stable pharmaceutical preparation useful for the treatment of pain, especially severe pain, without causing side-effects comprises oxycodone and naxolone in retarded release formulation
ES2546010T3 (en) 2002-04-05 2015-09-17 Euro-Celtique S.A. Pharmaceutical preparation containing oxycodone and naloxone
US20030191147A1 (en) 2002-04-09 2003-10-09 Barry Sherman Opioid antagonist compositions and dosage forms
US7670612B2 (en) 2002-04-10 2010-03-02 Innercap Technologies, Inc. Multi-phase, multi-compartment capsular delivery apparatus and methods for using same
EP1496931A4 (en) 2002-04-19 2009-07-01 Bristol Myers Squibb Co Methods for treating an autoimmune disease using a soluble ctla4 molecule and a dmard or nsaid
US20030199439A1 (en) 2002-04-22 2003-10-23 Simon David Lew Compositions of alpha3beta4 receptor antagonists and opioid agonist analgesics
US20030199496A1 (en) 2002-04-22 2003-10-23 Simon David Lew Pharmaceutical compositions containing alpha3beta4 nicotinic receptor antagonists and methods of their use
FR2838647B1 (en) 2002-04-23 2006-02-17 PROLONGED RELEASE PARTICLES, PROCESS FOR THEIR PREPARATION AND TABLETS CONTAINING SAME
US20040009172A1 (en) 2002-04-26 2004-01-15 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20030203027A1 (en) 2002-04-26 2003-10-30 Ethicon, Inc. Coating technique for deposition of drug substance on a substrate
US20050106249A1 (en) 2002-04-29 2005-05-19 Stephen Hwang Once-a-day, oral, controlled-release, oxycodone dosage forms
DE60325715D1 (en) 2002-04-29 2009-02-26 Alza Corp METHODS AND PHARMACEUTICAL FOR THE CONTROLLED RELEASE OF OXYCODONE
US7485298B2 (en) 2002-05-23 2009-02-03 Michael Powell Diagnosis and treatment of human dormancy-related sequellae
RU2004134728A (en) 2002-05-31 2005-06-10 Алза Корпорейшн (Us) DOSED FORMS AND COMPOSITIONS FOR THE OSMOTIC DELIVERY OF VARIOUS DOSAGE OXYCODONE
US8557291B2 (en) 2002-07-05 2013-10-15 Collegium Pharmaceutical, Inc. Abuse-deterrent pharmaceutical compositions of opioids and other drugs
US8840928B2 (en) 2002-07-05 2014-09-23 Collegium Pharmaceutical, Inc. Tamper-resistant pharmaceutical compositions of opioids and other drugs
US7985422B2 (en) 2002-08-05 2011-07-26 Torrent Pharmaceuticals Limited Dosage form
MXPA05001826A (en) 2002-08-15 2005-04-19 Euro Celtique Sa Pharmaceutical compositions.
US20050152905A1 (en) 2002-08-22 2005-07-14 Omoigui Osemwota S. Method of biochemical treatment of persistent pain
US20050020613A1 (en) 2002-09-20 2005-01-27 Alpharma, Inc. Sustained release opioid formulations and method of use
JP5189242B2 (en) 2002-09-23 2013-04-24 アルケルメス ファーマ アイルランド リミテッド Abuse-resistant pharmaceutical composition
US20040062778A1 (en) 2002-09-26 2004-04-01 Adi Shefer Surface dissolution and/or bulk erosion controlled release compositions and devices
US20040121010A1 (en) 2002-10-25 2004-06-24 Collegium Pharmaceutical, Inc. Pulsatile release compositions of milnacipran
CA2503381A1 (en) 2002-10-25 2004-05-13 Collegium Pharmaceutical, Inc. Stereoisomers of p-hydroxy-milnacipran, and methods of use thereof
US20070292461A1 (en) 2003-08-04 2007-12-20 Foamix Ltd. Oleaginous pharmaceutical and cosmetic foam
US7192966B2 (en) 2002-11-15 2007-03-20 Branded Products For The Future Pharmaceutical composition
US20040110781A1 (en) 2002-12-05 2004-06-10 Harmon Troy M. Pharmaceutical compositions containing indistinguishable drug components
US7670627B2 (en) 2002-12-09 2010-03-02 Salvona Ip Llc pH triggered targeted controlled release systems for the delivery of pharmaceutical active ingredients
CA2507522C (en) 2002-12-13 2015-02-24 Durect Corporation Oral drug delivery system
US20040224020A1 (en) 2002-12-18 2004-11-11 Schoenhard Grant L. Oral dosage forms with therapeutically active agents in controlled release cores and immediate release gelatin capsule coats
WO2004062577A2 (en) 2003-01-03 2004-07-29 Shire Laboratories Inc. Two or more enteric materials to regulate drug release
US7763043B2 (en) 2003-01-09 2010-07-27 Boston Scientific Scimed, Inc. Dilatation catheter with enhanced distal end for crossing occluded lesions
US20040259948A1 (en) 2003-01-10 2004-12-23 Peter Tontonoz Reciprocal regulation of inflammation and lipid metabolism by liver X receptors
CA2513432A1 (en) 2003-01-22 2004-08-05 Pfizer Products Inc. Methods for treating joint pain or improving sleep using an estrogen agonist/antagonist
US20060153915A1 (en) 2003-01-23 2006-07-13 Amorepacific Corporation Sustained-release preparations and method for producing the same
FR2850576B1 (en) 2003-02-05 2007-03-23 Ethypharm Sa COMPOSITION COMPRISING A MIXTURE OF ACTIVE INGREDIENTS AND PROCESS FOR PREPARING THE SAME
EP1605916A4 (en) 2003-02-12 2012-02-22 R & P Korea Co Ltd Solvent system of hardly soluble drug with improved elution rate
WO2004075832A2 (en) 2003-02-27 2004-09-10 Lucas John M Methods and compositions for the treatment of chronic pain using dhea and derivatives thereof
US20050004098A1 (en) 2003-03-20 2005-01-06 Britten Nancy Jean Dispersible formulation of an anti-inflammatory agent
SI1608407T1 (en) 2003-03-20 2006-12-31 Pharmacia Corp Dispersible formulation of an anti-inflammatory agent
ES2360102T3 (en) 2003-03-26 2011-05-31 Egalet A/S SYSTEM FOR CONTROLLED RELEASE OF MORPHINE.
CN1819819B (en) 2003-05-07 2011-03-09 株式会社三养社 Highly plastic granules for making fast melting tablets
WO2005123192A2 (en) 2004-06-17 2005-12-29 Osteologix A/S Improving pain treatment with strontium combinations
US8906413B2 (en) 2003-05-12 2014-12-09 Supernus Pharmaceuticals, Inc. Drug formulations having reduced abuse potential
US8029822B2 (en) 2003-05-22 2011-10-04 Osmotica Kereskedelmi és Seolgáltató KFT Rupturing controlled release device having a preformed passageway
JPWO2004108163A1 (en) 2003-06-04 2006-07-20 塩野義製薬株式会社 Method for producing low-chargeable powder
WO2005000331A2 (en) 2003-06-04 2005-01-06 Mucosal Therapeutics, Inc. Compositions for the treatment and prevention of degenerative joint disorders
US20040265378A1 (en) 2003-06-25 2004-12-30 Yingxu Peng Method and compositions for producing granules containing high concentrations of biologically active substances
TWI357815B (en) 2003-06-27 2012-02-11 Euro Celtique Sa Multiparticulates
US20060165790A1 (en) 2003-06-27 2006-07-27 Malcolm Walden Multiparticulates
WO2005004989A2 (en) 2003-07-01 2005-01-20 Todd Maibach Film comprising therapeutic agents
US20050074425A1 (en) 2003-07-02 2005-04-07 Polycord, Inc. Method for delivering polymerized therapeutic agent compositions and compositions thereof
CN1826100B (en) 2003-07-17 2010-12-22 旗帜药物胶囊公司 Controlled release preparation
US8075872B2 (en) 2003-08-06 2011-12-13 Gruenenthal Gmbh Abuse-proofed dosage form
DE10336400A1 (en) 2003-08-06 2005-03-24 Grünenthal GmbH Anti-abuse dosage form
US20070048228A1 (en) 2003-08-06 2007-03-01 Elisabeth Arkenau-Maric Abuse-proofed dosage form
DE102005005446A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Break-resistant dosage forms with sustained release
US20050059023A1 (en) 2003-09-16 2005-03-17 Cantor Thomas L. Methods and kits for monitoring resistance to therapeutic agents
PL1675622T3 (en) 2003-09-17 2017-11-30 Nektar Therapeutics Multi-arm polymer prodrugs
US20060194826A1 (en) 2003-09-25 2006-08-31 Euro-Celtique S.A. Pharmaceutical combinations of hydrocodone and naltrexone
SI1663229T1 (en) 2003-09-25 2010-08-31 Euro Celtique Sa Pharmaceutical combinations of hydrocodone and naltrexone
AU2004275835B2 (en) 2003-09-26 2011-06-23 Alza Corporation Controlled release formulations exhibiting an ascending rate of release
US20050158382A1 (en) 2003-09-26 2005-07-21 Evangeline Cruz Controlled release formulations of opioid and nonopioid analgesics
US20080031901A1 (en) 2004-09-24 2008-02-07 Abbott Laboratories Sustained release monoeximic formulations of opioid and nonopioid analgesics
WO2005038049A2 (en) 2003-10-06 2005-04-28 Heinrich Guenther System and method for optimizing drug therapy
US20060009478A1 (en) 2003-10-15 2006-01-12 Nadav Friedmann Methods for the treatment of back pain
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
EP1689367A1 (en) 2003-10-21 2006-08-16 Actavis Group HF Pharmaceutical formulations containing quetiapine
JP2007522079A (en) 2003-10-31 2007-08-09 エラン ファーマ インターナショナル リミテッド Nimesulide composition
EP1684732A2 (en) 2003-11-12 2006-08-02 Ranbaxy Laboratories Limited Ibuprofen-containing soft gelatin capsules
US7201920B2 (en) 2003-11-26 2007-04-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of opioid containing dosage forms
ATE473736T1 (en) 2003-12-16 2010-07-15 United Therapeutics Corp USE OF TREPROSTINIL FOR THE TREATMENT OF ISCHEMIC LESIONS
US20090124697A1 (en) 2003-12-16 2009-05-14 United Therapeutics Corporation Inhalation formulations of treprostinil
US20050266032A1 (en) 2003-12-17 2005-12-01 Sovereign Pharmaceuticals, Ltd. Dosage form containing multiple drugs
KR20060123493A (en) 2003-12-23 2006-12-01 알자 코포레이션 Methods and dosage forms for increasing solubility of drug compositions for controlled delivery
WO2005063219A2 (en) 2003-12-23 2005-07-14 Ranbaxy Laboratories Limited Ibuprofen-containing soft gelatin capsules
CA2551807A1 (en) 2004-01-14 2005-08-04 Gilead Sciences, Inc. Lipid-based dispersions useful for drug delivery
TWI350762B (en) 2004-02-12 2011-10-21 Euro Celtique Sa Particulates
US9016221B2 (en) 2004-02-17 2015-04-28 University Of Florida Research Foundation, Inc. Surface topographies for non-toxic bioadhesion control
WO2005092306A1 (en) 2004-03-11 2005-10-06 The General Hospital Corporation Methods and compositions for modulating opioid tolerance and chronic pain
US20070042969A1 (en) 2004-03-26 2007-02-22 Srz Properties, Inc. Combination therapy for pain in painful diabetic neuropathy
ES2244326B1 (en) 2004-04-05 2007-02-16 Laboratorios Del Dr. Esteve, S.A. COMBINATION OF ACTIVE SUBSTANCES.
US20050226929A1 (en) 2004-04-12 2005-10-13 Jianbo Xie Controlled release opioid analgesic formulation
GB0408308D0 (en) 2004-04-14 2004-05-19 Vectura Ltd Pharmaceutical compositions
WO2005102338A1 (en) 2004-04-20 2005-11-03 Pfizer Limited Method of treating neuropathic pain using a crth2 receptor antagonist
GB0408854D0 (en) 2004-04-21 2004-05-26 Univ York Separating method
WO2005107467A2 (en) 2004-05-03 2005-11-17 Descartes Therapeutics, Inc. Compositions including opioids and methods of their use in treating pain
US8216610B2 (en) 2004-05-28 2012-07-10 Imaginot Pty Ltd. Oral paracetamol formulations
US20050265955A1 (en) 2004-05-28 2005-12-01 Mallinckrodt Inc. Sustained release preparations
EP1758653A2 (en) 2004-06-17 2007-03-07 Osteologix A/S Treatments comprising strontium for rheumatic and arthritic diseases and pain
DE102004032049A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
US20060008527A1 (en) 2004-07-09 2006-01-12 Yury Lagoviyer Controlled phase composition technology as an improved process for protection of drugs
US20060039865A1 (en) 2004-07-26 2006-02-23 Preston David M Pharmaceutical compositions and methods for the prevention of drug misuse
US20060024368A1 (en) 2004-07-30 2006-02-02 Reza Fassihi Compressed composite delivery system for release-rate modulation of bioactives
US20070003622A1 (en) 2004-12-16 2007-01-04 Sovereign Pharmaceuticals, Ltd. Diphenhydramine containing dosage form
PT1789067E (en) 2004-08-12 2012-08-06 Helsinn Healthcare Sa Use of growth hormone secretagogues for stimulating the motility of the gastrointestinal system
US8039456B2 (en) 2004-08-12 2011-10-18 Helsinn Therapeutics (U.S.), Inc. Method of stimulating the motility of the gastrointestinal system using ipamorelin
US20060052370A1 (en) 2004-08-24 2006-03-09 Meyerson Laurence R Methods and compositions for treating nociceptive pain
GB2418854B (en) 2004-08-31 2009-12-23 Euro Celtique Sa Multiparticulates
WO2006030402A2 (en) 2004-09-17 2006-03-23 Ranbaxy Laboratories Limited Dual compartment osmotic delivery device
US20060062734A1 (en) 2004-09-20 2006-03-23 Melker Richard J Methods and systems for preventing diversion of prescription drugs
US8541026B2 (en) 2004-09-24 2013-09-24 Abbvie Inc. Sustained release formulations of opioid and nonopioid analgesics
US20060067971A1 (en) 2004-09-27 2006-03-30 Story Brooks J Bone void filler
US20060074422A1 (en) 2004-09-27 2006-04-06 Story Brooks J Suture anchor and void filler combination
EP3173072A1 (en) 2004-10-01 2017-05-31 Ramscor, Inc. Conveniently implantable sustained release drug compositions
AU2005294382A1 (en) 2004-10-04 2006-04-20 Qlt Usa, Inc. Ocular delivery of polymeric delivery formulations
WO2006044645A2 (en) 2004-10-13 2006-04-27 Adolor Corporation Sulfamoyl benzamides and methods of their use
WO2006046114A2 (en) 2004-10-25 2006-05-04 Ranbaxy Laboratories Limited Osmotic dosage forms providing ascending drug release, and processes for their preparation
CA2585471A1 (en) 2004-11-01 2006-05-11 Seo Hong Yoo Methods and compositions for reducing neurodegeneration in amyotrophic lateral sclerosis
US20060099254A1 (en) 2004-11-02 2006-05-11 Desai Divyakant S Sustained release drug delivery system and method
JP2006150061A (en) 2004-11-04 2006-06-15 Nec Corp Drug delivery system, and medicine capsule and signal transmitter used for the same
US20060111307A1 (en) 2004-11-16 2006-05-25 Wendye Robbins Methods and compositions for treating pain
US20070087977A1 (en) 2004-11-16 2007-04-19 Wendye Robbins Methods and compositions for treating pain
FR2878161B1 (en) 2004-11-23 2008-10-31 Flamel Technologies Sa ORAL MEDICINE FORM, SOLID AND DESIGNED TO AVOID MEASUREMENT
US20060177380A1 (en) 2004-11-24 2006-08-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
FR2878158B1 (en) 2004-11-24 2009-01-16 Flamel Technologies Sa ORAL PHARMACEUTICAL FORM, SOLID MICROPARTICULAR DESIGNED TO PREVENT MEASUREMENT
US20060110327A1 (en) 2004-11-24 2006-05-25 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US20080152595A1 (en) 2004-11-24 2008-06-26 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US20070231268A1 (en) 2004-11-24 2007-10-04 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US8574626B2 (en) 2004-12-03 2013-11-05 Osmotica Kereskedelmi és Szolgáltató KFT Osmotic device containing amantadine and an osmotic salt
CA2590802A1 (en) 2004-12-20 2006-06-29 Collegium Pharmaceutical, Inc. Pharmaceutical compositions for sleep disorders
MY146381A (en) 2004-12-22 2012-08-15 Amgen Inc Compositions and methods relating relating to anti-igf-1 receptor antibodies
ES2343641T3 (en) 2005-01-14 2010-08-05 Camurus Ab TOPIC BIOADHESIVE FORMULATIONS.
EP1848403B8 (en) 2005-01-14 2010-05-19 Camurus Ab Topical bioadhesive formulations
FR2881652B1 (en) 2005-02-08 2007-05-25 Flamel Technologies Sa MICROPARTICULAR ORAL PHARMACEUTICAL FORM ANTI-MEASURING
EP1845946A2 (en) 2005-02-10 2007-10-24 Orexo AB Pharmaceutical compositions useful in the transmucosal administration of drugs
BRPI0607402A2 (en) 2005-03-01 2009-09-01 Pfizer Ltd use of pde7 inhibitors to treat neuropathic pain
US20060205752A1 (en) 2005-03-14 2006-09-14 Keith Whitehead Stabilized hydrocodone pharmaceutical compositions with ethylenediaminetetraacetic acid
WO2006099541A2 (en) 2005-03-15 2006-09-21 Richard Daniel Carliss Therapeutic wound care product
FR2883179B1 (en) 2005-03-18 2009-04-17 Ethypharm Sa COATED TABLET
US9149439B2 (en) 2005-03-21 2015-10-06 Sandoz Ag Multi-particulate, modified-release composition
CN101151021A (en) 2005-03-28 2008-03-26 奥瑞克索股份公司 New pharmaceutical compositions useful in the treatment of pain
WO2006105205A1 (en) 2005-03-29 2006-10-05 University Of Kentucky Research Foundation Sublingual spray for the treatment of pain
WO2006103551A1 (en) 2005-03-31 2006-10-05 Ranbaxy Laboratories Limited Controlled release formulations of oxycodone
EP1898895A2 (en) 2005-04-22 2008-03-19 Wyeth Treatment of drug abuse
US9161918B2 (en) 2005-05-02 2015-10-20 Adare Pharmaceuticals, Inc. Timed, pulsatile release systems
EP1881819A1 (en) 2005-05-10 2008-01-30 Novartis AG Extrusion process for making compositions with poorly compressible therapeutic compounds
US20060263429A1 (en) 2005-05-20 2006-11-23 Hengsheng Feng Compressible mixture, compressed pharmaceutical compositions, and method of preparation thereof
US20090143761A1 (en) 2005-06-03 2009-06-04 Transdermal Patents Company, Llc Agent delivery system and uses of same
WO2007053197A2 (en) 2005-06-03 2007-05-10 Elan Pharma International, Limited Nanoparticulate acetaminophen formulations
US9060950B2 (en) 2005-06-13 2015-06-23 Paul H. Rosenberg, Proximate Concepts, LLC. Emetic embedded capsule
EP1893183A4 (en) 2005-06-13 2010-08-18 Paul Rosenberg Emetic embedded capsule
WO2006133733A1 (en) 2005-06-13 2006-12-21 Flamel Technologies Oral dosage form comprising an antimisuse system
US20070020339A1 (en) 2005-07-20 2007-01-25 Pharmorx Inc. Compositions and methods for controlling abuse of medications
WO2007021970A2 (en) 2005-08-15 2007-02-22 Praecis Pharmaceuticals, Inc. Stable pharmaceutical formulations and methods of use thereof
WO2007025182A2 (en) 2005-08-26 2007-03-01 Bpsi Holdings, Inc. Drug compositions containing controlled release hypromellose matrices
US8343546B2 (en) 2005-09-13 2013-01-01 Coating Place, Inc. Ion exchange resin treated to control swelling
KR100708713B1 (en) 2005-09-24 2007-04-17 삼성에스디아이 주식회사 Nanocomposite, naocomposite electrolyte membrane, and fuel cell using the same
FR2891459B1 (en) 2005-09-30 2007-12-28 Flamel Technologies Sa MICROPARTICLES WITH MODIFIED RELEASE OF AT LEAST ONE ACTIVE INGREDIENT AND ORAL GALENIC FORM COMPRISING THE SAME
WO2007050631A2 (en) 2005-10-25 2007-05-03 Cima Labs Inc. Dosage form with coated active
WO2007050975A2 (en) 2005-10-26 2007-05-03 Banner Pharmacaps, Inc. Hydrophilic vehicle-based dual controlled release matrix system as capsule fill
PL116330U1 (en) 2005-10-31 2007-04-02 Alza Corp Method for the reduction of alcohol provoked rapid increase in the released dose of the orally administered opioide with prolonged liberation
US20100210732A1 (en) 2005-11-02 2010-08-19 Najib Babul Methods of Preventing the Serotonin Syndrome and Compositions for Use Therefor
WO2008134071A1 (en) 2007-04-26 2008-11-06 Theraquest Biosciences, Inc. Multimodal abuse resistant extended release formulations
WO2007087452A2 (en) 2006-01-27 2007-08-02 Theraquest Biosciences, Llc Abuse resistant and extended release formulations and method of use thereof
US8652529B2 (en) 2005-11-10 2014-02-18 Flamel Technologies Anti-misuse microparticulate oral pharmaceutical form
US7544676B2 (en) 2005-11-10 2009-06-09 Adolor Corporation Sulfamoyl benzamides and methods of their use
WO2007132293A2 (en) 2005-11-10 2007-11-22 Circ Pharma Research And Development Limited Once-daily administration of central nervous system drugs
FR2892937B1 (en) 2005-11-10 2013-04-05 Flamel Tech Sa MICROPARTICULAR ORAL PHARMACEUTICAL FORM ANTI-MEASURING
US20070134493A1 (en) 2005-12-08 2007-06-14 Kanji Meghpara Compositions and capsules with stable hydrophilic layers
BRPI0619806A2 (en) 2005-12-13 2011-10-18 Biodelivery Sciences Int Inc abuse resistant transmucosal drug delivery device
WO2007072503A2 (en) 2005-12-21 2007-06-28 Panacea Biotec Ltd. Combinations for managing inflammation and associated disorders
TW200734334A (en) 2006-01-13 2007-09-16 Wyeth Corp Treatment of substance abuse
US20100172989A1 (en) 2006-01-21 2010-07-08 Abbott Laboratories Abuse resistant melt extruded formulation having reduced alcohol interaction
US20090317355A1 (en) 2006-01-21 2009-12-24 Abbott Gmbh & Co. Kg, Abuse resistant melt extruded formulation having reduced alcohol interaction
US20090022798A1 (en) 2007-07-20 2009-01-22 Abbott Gmbh & Co. Kg Formulations of nonopioid and confined opioid analgesics
JP2009523833A (en) * 2006-01-21 2009-06-25 アボット ゲーエムベーハー ウント カンパニー カーゲー Formulations and methods for drug delivery
EP1813276A1 (en) 2006-01-27 2007-08-01 Euro-Celtique S.A. Tamper resistant dosage forms
EP1976492B8 (en) 2006-01-27 2018-07-04 Adare Pharmaceuticals, Inc. Drug delivery systems comprising weakly basic drugs and organic acids
GB0603008D0 (en) 2006-02-14 2006-03-29 Portela & Ca Sa Method
US20070213822A1 (en) 2006-02-14 2007-09-13 Sdgi Holdings, Inc. Treatment of the vertebral column
US20070213717A1 (en) 2006-02-14 2007-09-13 Sdgi Holdings, Inc. Biological fusion in the vertebral column
US20070213718A1 (en) 2006-02-14 2007-09-13 Sdgi Holdings, Inc. Treatment of the vertebral column
FR2897267A1 (en) 2006-02-16 2007-08-17 Flamel Technologies Sa MULTIMICROPARTICULAR PHARMACEUTICAL FORMS FOR PER OS ADMINISTRATION
US8940796B2 (en) 2006-02-21 2015-01-27 Wyeth Llc Phenylephrine liquid formulations
AU2007223560A1 (en) 2006-03-01 2007-09-13 Tristrata, Inc. Composition and method for topical treatment of tar-responsive dermatological disorders
ZA200807571B (en) 2006-03-01 2009-08-26 Ethypharm Sa Crush-resistant tablets intended to prevent accidental misuse and unlawful diversion
JP5349059B2 (en) 2006-03-06 2013-11-20 ポーゼン インコーポレイテッド Dosage form for administering a combination of drugs
US9265732B2 (en) 2006-03-06 2016-02-23 Pozen Inc. Dosage forms for administering combinations of drugs
US20070212417A1 (en) 2006-03-07 2007-09-13 Cherukuri S R Compressible resilient granules and formulations prepared therefrom
US20080069889A1 (en) 2006-03-07 2008-03-20 Cherukuri S R Compressible resilient granules and formulations prepared therefrom
EP1993519A4 (en) 2006-03-15 2011-12-21 Acura Pharmaceuticals Inc Methods and compositions for deterring abuse of orally administered pharmaceutical products
BRPI0709606B8 (en) 2006-03-16 2021-05-25 Tris Pharma Inc orally administrable liquid suspension with modified release characteristics
CN101453993A (en) 2006-04-03 2009-06-10 伊萨·奥迪迪 Controlled release delivery device comprising an organosol coat
US10022339B2 (en) 2006-04-21 2018-07-17 The Procter & Gamble Company Compositions and methods useful for treatment of respiratory illness
EP2015740A4 (en) 2006-04-24 2009-11-11 Panacea Biotec Ltd Novel low dose pharmaceutical compositions comprising nimesulide, preparation and use thereof
US20070254027A1 (en) 2006-04-28 2007-11-01 The Procter & Gamble Company Compositions and methods useful for treatment of respiratory illness
ES2626632T3 (en) 2006-05-09 2017-07-25 Mallinckrodt Llc Solid dosage forms of zero-order modified release
EP2021502A4 (en) 2006-05-09 2010-08-25 Mas Metabolic Analytical Servi Novel genes and markers in type 2 diabetes and obesity
US8377994B2 (en) 2006-05-10 2013-02-19 Evonik Degussa GmeH Use of roll compacted pyrogenically produced silicon dioxide in pharmaceutical compositions
US10960077B2 (en) 2006-05-12 2021-03-30 Intellipharmaceutics Corp. Abuse and alcohol resistant drug composition
US9023400B2 (en) 2006-05-24 2015-05-05 Flamel Technologies Prolonged-release multimicroparticulate oral pharmaceutical form
FR2901478B1 (en) 2006-05-24 2015-06-05 Flamel Tech Sa MULTIMICROPARTICULATED ORAL PHARMACEUTICAL FORM WITH PROLONGED RELEASE
US20070292508A1 (en) 2006-06-05 2007-12-20 Balchem Corporation Orally disintegrating dosage forms
US20070281016A1 (en) 2006-06-06 2007-12-06 Endo Pharmaceuticals Inc., A Delaware Corporation Sustained release oxycodone composition with acrylic polymer and surfactant
US20070281017A1 (en) 2006-06-06 2007-12-06 Endo Pharmaceuticals Inc., A Delaware Corporation Sustained release oxycodone composition with acrylic polymer and metal hydroxide
US20070286900A1 (en) 2006-06-09 2007-12-13 Catherine Herry Low dose tablets of opioid analgesics and preparation process
US20080069891A1 (en) 2006-09-15 2008-03-20 Cima Labs, Inc. Abuse resistant drug formulation
KR20090038431A (en) 2006-06-19 2009-04-20 맥네일-피피씨, 인코포레이티드 Enteric coated particles containing an active ingredient
PL2526932T3 (en) 2006-06-19 2017-12-29 Alpharma Pharmaceuticals Llc Pharmaceutical composition
EP2032124A1 (en) 2006-06-27 2009-03-11 Biovail Laboratories International Srl Multiparticulate osmotic delivery system
GB0613925D0 (en) 2006-07-13 2006-08-23 Unilever Plc Improvements relating to nanodispersions
EP2043613A1 (en) 2006-07-14 2009-04-08 Fmc Corporation Solid form
US20080014274A1 (en) 2006-07-14 2008-01-17 Wyeth Enhanced stability phenylephrine liquid compositions
US8765178B2 (en) 2006-07-19 2014-07-01 Watson Laboratories, Inc. Controlled release formulations and associated methods
WO2008011596A2 (en) 2006-07-21 2008-01-24 Lab International Srl Hydrophilic abuse deterrent delivery system
US9005652B2 (en) 2006-07-25 2015-04-14 Wyeth Chewable tablet containing phenylephrine
WO2008013416A1 (en) 2006-07-27 2008-01-31 Amorepacific Corporation Process for preparing powder comprising nanoparticles of sparingly soluble drug
US20080026040A1 (en) 2006-07-31 2008-01-31 Isaac Farr Active agent-releasing dosage forms
US20080026062A1 (en) 2006-07-31 2008-01-31 Isaac Farr Pharmaceutical compositions including nano-sized active agent
KR101445757B1 (en) 2006-08-04 2014-10-02 에씨팜 Granule and orally disintegrating tablet comprising oxycodone
ATE462416T1 (en) 2006-08-04 2010-04-15 Ethypharm Sa MULTI-LAYER OROUGH DISSOLVING TABLET
SA07280459B1 (en) 2006-08-25 2011-07-20 بيورديو فارما إل. بي. Tamper Resistant Oral Pharmaceutical Dosage Forms Comprising an Opioid Analgesic
WO2008027442A2 (en) 2006-08-30 2008-03-06 Theraquest Biosciences, Llc Abuse deterrent oral pharmaceutical formulations of opioid agonists and method of use
US20080057122A1 (en) 2006-08-31 2008-03-06 Aaipharma Inc. Acetaminophen pharmaceutical compositions
RU2434630C2 (en) 2006-08-31 2011-11-27 Юранд, Инк. Systems of medication delivery, including solid solutions of weak-base medications
WO2008033351A2 (en) 2006-09-11 2008-03-20 Theraquest Biosciences, Inc. Multimodal abuse resistant and extended release formulations
US8445018B2 (en) 2006-09-15 2013-05-21 Cima Labs Inc. Abuse resistant drug formulation
KR20080026754A (en) 2006-09-21 2008-03-26 주식회사 삼양사 Release controlled particle comprising a biologically active substance, and preparing method thereof
DE102006044694A1 (en) 2006-09-22 2008-03-27 Krewel Meuselbach Gmbh Peroral solid analgesic preparation
WO2008042331A2 (en) 2006-09-29 2008-04-10 Monosol Rx Llc Film embedded packaging and method of making same
US20080085312A1 (en) 2006-10-04 2008-04-10 Auriga Laboratories, Inc. Multi-Phase Release Potassium Guaiacolsulfonate Compositions
US8653066B2 (en) 2006-10-09 2014-02-18 Charleston Laboratories, Inc. Pharmaceutical compositions
PT2124556E (en) 2006-10-09 2014-12-03 Charleston Lab Inc Pharmaceutical compositions
US20080085304A1 (en) 2006-10-10 2008-04-10 Penwest Pharmaceuticals Co. Robust sustained release formulations
CA2666582C (en) 2006-10-20 2014-09-09 Mcneil-Ppc, Inc. Acetaminophen / ibuprofen combinations
KR101441459B1 (en) 2006-10-24 2014-09-18 헬신 헬쓰케어 에스.에이. Soft capsules comprising palonosetron hydrochloride having improved stability and bioavailability
US20080102123A1 (en) 2006-10-27 2008-05-01 Schachter Deborah M Self-gelling tunable drug delivery system
MX2009004965A (en) 2006-11-07 2009-06-05 Nektar Therapeutics Al Corp Dosage forms and co-administration of an opioid agonist and an opioid antagonist.
US7842662B2 (en) 2006-11-10 2010-11-30 Cara Therapeutics, Inc. Synthetic peptide amide dimers
US20080293695A1 (en) 2007-05-22 2008-11-27 David William Bristol Salts of physiologically active and psychoactive alkaloids and amines simultaneously exhibiting bioavailability and abuse resistance
KR101513736B1 (en) 2006-11-10 2015-04-20 케러 테라퓨틱스, 인코포레이티드 Synthetic peptide amides
WO2008063625A2 (en) 2006-11-20 2008-05-29 Adolor Corporation Pyridine compounds and methods of their use
BRPI0719587A2 (en) 2006-11-22 2013-12-17 Progenics Pharm Inc (S) - 7,8-SATURATED-4,5-EPOXY-MORFINAN ANALOG STEREO ISOMERS
AU2007325315A1 (en) 2006-11-28 2008-06-05 Kalypsys Inc Heterocyclic modulators of TGR5
US20080132751A1 (en) 2006-11-30 2008-06-05 Betazone Laboratories, Inc. Compositions and methods for treatment of pain
PT2101740E (en) 2006-12-04 2013-12-23 Orexo Ab New non-abusable pharmaceutical composition comprising opioids
NZ577560A (en) 2007-01-16 2012-01-12 Egalet Ltd Use of i) a polyglycol and ii) an active drug substance for the preparation of a pharmaceutical composition for i) mitigating the risk of alcohol induced dose dumping and/or ii) reducing the risk of drug abuse
WO2008097976A1 (en) 2007-02-09 2008-08-14 Kalypsys, Inc. Heterocyclic modulators of tgr5 for treatment of disease
WO2008100977A2 (en) 2007-02-14 2008-08-21 N.V. Organon Carbamates therapeutic release agents as amidase inhibitors
EP2650303A1 (en) 2007-02-26 2013-10-16 Ironwood Pharmaceuticals, Inc. Methods and compositions for the treatment of heart failure and other disorders
US20100144591A1 (en) 2007-03-02 2010-06-10 Schering Corporation Benzimidazole derivatives and methods of use thereof
ATE440598T1 (en) 2007-03-02 2009-09-15 Flamek Corp Ou ANALGESIC COMPOSITION OF TOPICALLY APPLIED NON-STEROID AND ANTI-INFLAMMATORY DRUGS AND OPIOIDS
MX2009009417A (en) 2007-03-02 2009-09-11 Schering Corp Piperidinyl-piperidine and piperazinyl-piperidine for use in the treatment of diabetes or pain.
ES2707551T3 (en) 2007-03-02 2019-04-04 Amgen Inc Methods and compositions for treating tumor diseases
DE102007011485A1 (en) 2007-03-07 2008-09-11 Grünenthal GmbH Dosage form with more difficult abuse
US8173666B2 (en) 2007-03-12 2012-05-08 Nektar Therapeutics Oligomer-opioid agonist conjugates
US20080226702A1 (en) 2007-03-16 2008-09-18 Endo Pharmaceuticals, Inc. Transdermal Delivery Form Disposal Systems and Methods
TWI417099B (en) 2007-03-23 2013-12-01 Neuraxon Inc Quinolone and tetrahydroquinoline and related compounds having nos inhibitory activity
EP1980245A1 (en) 2007-04-11 2008-10-15 Cephalon France Bilayer lyophilized pharmaceutical compositions and methods of making and using same
US20100080829A1 (en) 2007-04-11 2010-04-01 Cephalon France Lyophilized pharmaceutical compositions and methods of making and using same
US20080260837A1 (en) 2007-04-20 2008-10-23 Qpharma, L.L.C. Physically stable aqueous suspensions of active pharmaceuticals
WO2008133928A2 (en) 2007-04-27 2008-11-06 The Gi Company, Inc. Mucin glycoproteins and their use for treatment of epithelial lesions and mucin dependent disorders
ES2855700T3 (en) 2007-04-27 2021-09-24 Cydex Pharmaceuticals Inc Formulations containing clopidogrel and sulfoalkyl ether-cyclodextrin and methods of use
JP2010526053A (en) 2007-05-01 2010-07-29 シグモイド・ファーマ・リミテッド Nimodipine pharmaceutical composition
DE102007021549A1 (en) 2007-05-08 2008-11-13 Novosis Ag Transdermal therapeutic system containing at least two opioids
WO2008141189A1 (en) 2007-05-09 2008-11-20 Elixir Pharmaceuticals, Inc. Ghrelin modulating compounds and combinations thereof
US20080286344A1 (en) 2007-05-16 2008-11-20 Olivia Darmuzey Solid form
WO2008140461A1 (en) 2007-05-16 2008-11-20 Fmc Corporation Solid form
US20080286343A1 (en) 2007-05-16 2008-11-20 Dzenana Cengic Solid form
WO2008140460A1 (en) 2007-05-16 2008-11-20 Fmc Corporation Solid form
WO2008140459A1 (en) 2007-05-16 2008-11-20 Fmc Corporation Solid form
US20080311162A1 (en) 2007-05-16 2008-12-18 Olivia Darmuzey Solid form
US20080292683A1 (en) 2007-05-24 2008-11-27 Monosolrx, Llc. Film shreds and delivery system incorporating same
EP2167048B1 (en) 2007-05-30 2016-10-26 Wockhardt Limited A novel tablet dosage form
US8202542B1 (en) 2007-05-31 2012-06-19 Tris Pharma Abuse resistant opioid drug-ion exchange resin complexes having hybrid coatings
AU2008258596B2 (en) 2007-06-04 2013-02-14 Egalet Ltd Controlled release pharmaceutical compositions for prolonged effect
CA2721135A1 (en) 2007-06-13 2008-12-24 Wayne State University Board Of Governors A zwitterion solution for low-volume therapeutic delivery
WO2008155620A1 (en) 2007-06-20 2008-12-24 Develco Pharma Schweiz Ag Dosage form containing dispersible matrix of sustained release granules
WO2009002299A1 (en) 2007-06-22 2008-12-31 Gluconova Llc Method for treating warm-blooded vertebrates with halide-free glucosamine-acidic drug complexes
US20090004281A1 (en) 2007-06-26 2009-01-01 Biovail Laboratories International S.R.L. Multiparticulate osmotic delivery system
US20090004285A1 (en) 2007-06-29 2009-01-01 Liangping Yu Stable non-disintegrating dosage forms and method of making same
EP2170051A1 (en) 2007-07-01 2010-04-07 Joseph Peter Habboushe Combination tablet with chewable outer layer
CA2690829A1 (en) 2007-07-20 2009-01-29 Abbott Gmbh & Co. Kg Formulations of nonopioid and confined opioid analgesics
WO2009018169A1 (en) 2007-07-27 2009-02-05 Auspex Pharmaceuticals, Inc. Substituted cyclohexanols
EP2182804B1 (en) 2007-08-06 2017-05-24 Biotie Therapies, Inc. Methods for treating dependence
CN105534936B (en) 2007-08-13 2020-07-31 Ohemo 生命科学股份有限公司 Abuse resistant drugs, methods of use and methods of preparation
US20090105124A1 (en) 2007-08-23 2009-04-23 Kalypsys, Inc. Heterocyclic modulators of tgr5
KR101545874B1 (en) 2007-09-03 2015-08-20 나노테라퓨틱스, 인코포레이티드 Particulate compositions for delivery of poorly soluble drugs
US20090068247A1 (en) 2007-09-12 2009-03-12 Mucosal Therapeutics Biocompatible devices coated with a tribonectin and methods for their production
KR20100087287A (en) 2007-09-17 2010-08-04 맥네일-피피씨, 인코포레이티드 Dip coated compositions containing copolymer of polyvinyl alcohol and polyethylene glycol and a gum
JP5204847B2 (en) 2007-09-21 2013-06-05 エボニック レーム ゲゼルシャフト ミット ベシュレンクテル ハフツング PH-dependent controlled release pharmaceutical opioid composition resistant to the effects of ethanol
US20090087483A1 (en) 2007-09-27 2009-04-02 Sison Raymundo A Oral dosage combination pharmaceutical packaging
KR20100082767A (en) 2007-10-01 2010-07-19 바스프 에스이 Rodenticide mixture
US20090110724A1 (en) 2007-10-31 2009-04-30 Everett Laboratories, Inc. Compositions and methods for treatment of pain
WO2009066146A2 (en) 2007-11-19 2009-05-28 Cadila Pharmaceuticals Ltd. Stable solutions of sparingly soluble actives
WO2009073686A1 (en) 2007-12-03 2009-06-11 Trans Dermal Patents Company, Llc Agent delivery system and uses of the same
CA2706658A1 (en) 2007-12-06 2009-06-18 Durect Corporation Methods useful for the treatment of pain, arthritic conditions or inflammation associated with a chronic condition
BRPI0820997A2 (en) 2007-12-10 2014-12-23 Eurand Inc Oral disintegration tablets comprising diphenhydramine.
JP5651818B2 (en) 2007-12-17 2015-01-14 パラディン ラブス インコーポレーテッド Controlled release formulation to prevent misuse
HUE029444T2 (en) 2008-01-04 2017-02-28 Schabar Res Ass Llc Composition comprising an analgesic and an antihistamine
EP2240022B1 (en) 2008-01-09 2016-12-28 Charleston Laboratories, Inc. Bilayered tablets comprising oxycodone and promethazine
US8497303B2 (en) 2008-01-18 2013-07-30 Dow Global Technologies Llc Method to enhance aqueous solubility of poorly soluble actives
EP2249811A1 (en) 2008-01-25 2010-11-17 Grünenthal GmbH Pharmaceutical dosage form
US9226907B2 (en) 2008-02-01 2016-01-05 Abbvie Inc. Extended release hydrocodone acetaminophen and related methods and uses thereof
MX2010008420A (en) 2008-02-01 2012-09-19 Abbott Lab Extended release hydrocodone acetaminophen and related methods and uses thereof.
US8088150B2 (en) 2008-02-04 2012-01-03 Aleeva Medical Inc. Device for disc shunt implantation and peri-shunt injection
KR101094231B1 (en) 2008-02-18 2011-12-14 하나제약 주식회사 Sustained release solid formulations and methods of manufacturing the same
EP2265287B1 (en) 2008-03-04 2018-09-05 Teva Pharmaceuticals International GmbH Methods of treating chronic pain
UY31698A (en) 2008-03-11 2009-11-10 Takeda Pharmaceutical SOLID PREPARATION OF ORAL DISINTEGRATION
US8372432B2 (en) 2008-03-11 2013-02-12 Depomed, Inc. Gastric retentive extended-release dosage forms comprising combinations of a non-opioid analgesic and an opioid analgesic
WO2009114648A1 (en) 2008-03-11 2009-09-17 Depomed Inc. Gastric retentive extended-release dosage forms comprising combinations of a non-opioid analgesic and an opioid analgesic
WO2009117130A2 (en) 2008-03-21 2009-09-24 Mylan Pharmaceuticals, Inc. Extended release forumulation containing a wax
US20090246265A1 (en) 2008-03-26 2009-10-01 Alltranz Inc. Abuse deterrent transdermal formulations of opiate agonists and agonist-antagonists
US8623401B2 (en) 2008-03-27 2014-01-07 Fenwafe Inc. Wafer formulation
WO2009118764A1 (en) 2008-03-28 2009-10-01 Panacea Biotec Limited Pharmaceutical composition comprising diclofenac and paracetamol
EP2106799A1 (en) 2008-03-31 2009-10-07 OWEN Holding LTD Use of a biologically active blood serum for the treatment of a disorder characterized in a reduced function of a GABA receptor
WO2009124755A1 (en) 2008-04-08 2009-10-15 European Molecular Biology Laboratory (Embl) Compounds with novel medical uses and method of identifying such compounds
WO2009126931A2 (en) 2008-04-11 2009-10-15 Xvasive, Inc. Combination therapy for bipolar disorder
JP5651291B2 (en) 2008-04-11 2015-01-07 株式会社センカファーマシー Polyethylene glycol derivative and process for producing the intermediate
US8524267B2 (en) 2008-04-18 2013-09-03 Warsaw Orthopedic, Inc. Dexamethasone formulations in a biodegradable material
US8420114B2 (en) 2008-04-18 2013-04-16 Warsaw Orthopedic, Inc. Alpha and beta adrenergic receptor agonists for treatment of pain and / or inflammation
ES2563061T3 (en) 2008-04-28 2016-03-10 Zogenix, Inc. New formulations for the treatment of migraine
CA2723307C (en) 2008-05-05 2017-08-22 Euro-Celtique S.A. Tamper resistant opioid composition for treating skin lesions
BRPI0912219A2 (en) 2008-05-07 2015-10-06 Nektar Therapeutics method, unit dose form, and peripherally acting orally administrable opioid antagonist.
RU2010151951A (en) 2008-05-20 2012-06-27 Ньюроджесэкс, Инк. (Us) GENERAL PROCEDURES OF HEPATOPROTECTOR AND ACETAMINOPHEN
CA2724877C (en) 2008-05-20 2016-09-13 Neurogesx, Inc. Water-soluble acetaminophen analogs
EP2123626A1 (en) 2008-05-21 2009-11-25 Laboratorios del Dr. Esteve S.A. Co-crystals of duloxetine and co-crystal formers for the treatment of pain
WO2009152133A1 (en) 2008-06-10 2009-12-17 Abbott Laboratories Novel tricyclic compounds
ZA200903858B (en) 2008-06-19 2013-10-30 Univ Of Witwatesrand Johannesburg Pharmaceutical dosage form
UA105182C2 (en) 2008-07-03 2014-04-25 Ньюрексон, Інк. Benzoxazines, benzothiazines, and related compounds having nos inhibitory activity
US20100003322A1 (en) 2008-07-03 2010-01-07 Lai Felix S Enteric coated hydrophobic matrix formulation
GB0813929D0 (en) 2008-07-30 2008-09-03 Glaxo Group Ltd Novel method
CA2734095A1 (en) 2008-08-14 2010-02-18 Bioneer A/S Coated tablets with a remaining degradation surface over the time
ES2562799T3 (en) 2008-08-19 2016-03-08 Adcock Ingram Intellectual Property (Pty) Limited Modulated speed administration of drugs from a three-layer tablet comprising tramadol, diclofenac, paracetamol
US20100076074A1 (en) 2008-08-26 2010-03-25 Auspex Pharmaceuticals, Inc. Carbamate reducers of skeletal muscle tension
JP5345813B2 (en) 2008-08-28 2013-11-20 テイカ製薬株式会社 Oxycodone transdermally absorbable pharmaceutical composition, pharmaceutical composition storage unit, and transdermally absorbable preparation using the same
US8067243B2 (en) 2008-09-03 2011-11-29 Oregon Medical Laboratories Methods and systems for analyzing medication levels in a sample
WO2010033195A1 (en) 2008-09-16 2010-03-25 Nektar Therapeutics Pegylated opioids with low potential for abuse
CA2737257C (en) 2008-09-18 2016-11-15 Purdue Pharma L.P. Pharmaceutical dosage forms comprising poly(.epsilon.-caprolactone)
CN102164589A (en) 2008-09-24 2011-08-24 赢创罗姆有限公司 PH-dependent controlled release pharmaceutical composition for non-opioids with resistance against the influence of ethanol
CA2737307A1 (en) 2008-10-14 2010-04-22 Mcneil Ab Multi portion intra-oral dosage form and use thereof
WO2010044842A1 (en) 2008-10-16 2010-04-22 University Of Tennessee Research Foundation Tamper resistant oral dosage forms containing an embolizing agent
US20100098746A1 (en) 2008-10-20 2010-04-22 Warsaw Orthopedic, Inc. Compositions and methods for treating periodontal disease comprising clonidine, sulindac and/or fluocinolone
RU2673882C1 (en) 2008-10-30 2018-12-03 Грюненталь Гмбх New and effective medicinal forms of tapentadol
US9072799B2 (en) 2008-10-31 2015-07-07 The Invention Science Fund I, Llc Compositions and methods for surface abrasion with frozen particles
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US20100266682A1 (en) 2008-12-10 2010-10-21 Nipun Davar Polyethylene glycol-coated sodium carbonate as a pharmaceutical excipient and compositions produced from the same
US20100143486A1 (en) 2008-12-10 2010-06-10 Nipun Davar Polyethylene glycol-coated sodium carbonate as a pharmaceutical excipient and compositions produced from the same
US8486449B2 (en) 2008-12-16 2013-07-16 Paladin Labs Inc. Misuse preventative, controlled release formulation
CA2750144C (en) 2008-12-31 2016-10-25 Upsher-Smith Laboratories, Inc. Opioid-containing oral pharmaceutical compositions and methods
US20100173013A1 (en) 2009-01-08 2010-07-08 Denis Drygin Treatment of neoplastic disorders using combination therapies
EP2389169A1 (en) 2009-01-26 2011-11-30 Egalet A/S Controlled release formulations with continuous efficacy
JP5408703B2 (en) 2009-01-30 2014-02-05 国立大学法人 千葉大学 Drug and production method thereof
EP2393487B1 (en) 2009-02-06 2016-11-02 Egalet Ltd. Pharmaceutical compositions resistant to abuse
US20110311626A1 (en) 2009-02-23 2011-12-22 Gopi Venkatesh Controlled release compositions comprising anti-cholinergic drugs
MX336355B (en) 2009-03-04 2016-01-14 Orexo Ab Abuse resistant formulation.
US20110311628A1 (en) 2009-03-09 2011-12-22 Council Of Scientific And Industrial Research Pulsatile release composition of therapeutic agent
US8945622B2 (en) 2009-03-09 2015-02-03 Council Of Scientific And Industrial Research Sustained release composition of therapeutic agent
SG174286A1 (en) 2009-03-10 2011-10-28 Euro Celtique Sa Immediate release pharmaceutical compositions comprising oxycodone and naloxone
SI2408436T1 (en) 2009-03-18 2017-06-30 Evonik Roehm Gmbh Controlled release pharmaceutical composition with resistance against the influence of ethanol employing a coating comprising neutral vinyl polymers and excipients
US20110311631A1 (en) 2009-03-18 2011-12-22 Evonik Röhm Gmbh Controlled release pharmaceutical composition with resistance against the influence of ethanol employing a coating comprising a polymer mixture and excipients
RU2589823C2 (en) 2009-04-09 2016-07-10 Алкермес Фарма Айэленд Лимитед Drug delivery composition
CN102481370A (en) 2009-04-17 2012-05-30 阿克西亚制药有限责任公司 Polymeric drug delivery systems and processes for producing such systems
WO2010123999A2 (en) 2009-04-21 2010-10-28 Auspex Pharmaceuticals, Inc. 1-methylpyrazole modulators of substance p, calcitonin gene-related peptide, adrenergic receptor, and/or 5-ht receptor
WO2010124089A2 (en) 2009-04-22 2010-10-28 Synosia Therapeutics, Inc. Methods for treating dependence
AU2010242749B2 (en) 2009-05-01 2015-05-07 Adare Pharmaceuticals, Inc. Orally disintegrating tablet compositions comprising combinations of non-opioid and opioid analgesics
US11135188B2 (en) 2009-05-11 2021-10-05 Bayer Healthcare Llc Method and composition to improve absorption of therapeutic agents
US20100291201A1 (en) 2009-05-14 2010-11-18 Cerovene, Inc. Coated pharmaceutical capsule dosage form
BRPI1009072B1 (en) 2009-05-18 2021-06-22 Adamis Pharmaceuticals Corporation DEVICE FOR DRY POWDER INHALATION AND DEVICE FOR DRY POWDER STORAGE
WO2010138441A1 (en) 2009-05-28 2010-12-02 Aptapharma, Inc. Multilayer oral tablets containing a non-steroidal anti-inflammatory drug and/or acetaminophen
WO2010141505A1 (en) 2009-06-01 2010-12-09 Protect Pharmaceutical Corporation Abuse-resistant delivery systems
US20100304998A1 (en) 2009-06-02 2010-12-02 Marquette University Chemical Proteomic Assay for Optimizing Drug Binding to Target Proteins
GB0909680D0 (en) 2009-06-05 2009-07-22 Euro Celtique Sa Dosage form
FR2946533A1 (en) 2009-06-12 2010-12-17 Ethypharm Sa REDUCTION OF PLASMATIC FLUCTUATIONS OF OPIOIDS.
EP2445487A2 (en) 2009-06-24 2012-05-02 Egalet Ltd. Controlled release formulations
AU2010263264B2 (en) 2009-06-25 2015-02-12 Certusview Technologies, Llc Locating equipment for and methods of simulating locate operations for training and/or skills evaluation
WO2010150930A1 (en) 2009-06-25 2010-12-29 (주)벡스코아 Fast dissolving film for oral administration which prevents unpleasant taste effectively
SG176897A1 (en) 2009-06-25 2012-01-30 Savient Pharmaceuticals Inc Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
KR101074271B1 (en) 2009-06-25 2011-10-17 (주)차바이오앤디오스텍 Fast dissolving oral dosage form containing steviosides as a taste masking agent
US20110003005A1 (en) 2009-07-06 2011-01-06 Gopi Venkatesh Methods of Treating PDNV and PONV with Extended Release Ondansetron Compositions
WO2011006012A1 (en) 2009-07-08 2011-01-13 Charleston Laboratories Inc. Pharmaceutical compositions
WO2011008298A2 (en) 2009-07-16 2011-01-20 Nectid, Inc. Novel axomadol dosage forms
EP2456469A1 (en) 2009-07-21 2012-05-30 Nektar Therapeutics Oligomer-opioid agonist conjugates
EP2456427B1 (en) 2009-07-22 2015-03-04 Grünenthal GmbH Hot-melt extruded controlled release dosage form
US20110020423A1 (en) 2009-07-22 2011-01-27 Puretech Ventures Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
PE20120572A1 (en) 2009-07-22 2012-06-06 Gruenenthal Chemie HANDLING RESISTANT STABILIZED OXIDATION DOSAGE FORM
NZ597960A (en) 2009-07-31 2013-07-26 Ascendis Pharma As Biodegradable polyethylene glycol based water-insoluble hydrogels
AU2010286354A1 (en) 2009-08-31 2012-04-19 Depomed, Inc. Gastric retentive pharmaceutical compositions for immediate and extended release of acetaminophen
MX2012003082A (en) 2009-09-17 2012-04-19 Cadila Healthcare Ltd Pharmaceutical compositions for reducing alcohol-induced dose dumping.
AU2010300641B2 (en) 2009-09-30 2016-03-17 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse
FR2951378B1 (en) 2009-10-16 2012-06-01 Flamel Tech Sa ANTI-MEASURING SOLID ORAL PHARMACEUTICAL FORM WITH A SPECIFIC MODIFIED RELEASE PROFILE
US20110104272A1 (en) 2009-11-05 2011-05-05 Depomed, Inc. Gastric retentive extended-release dosage forms comprising combinations of acetaminophen and phenylephrine
WO2011057199A1 (en) 2009-11-06 2011-05-12 Adenios, Inc. Compositions for treating cns disorders
CA2782177C (en) 2009-11-30 2019-10-15 Aptalis Pharmatech, Inc. Compressible-coated pharmaceutical compositions and tablets and methods of manufacture
EP2506716B1 (en) 2009-12-01 2017-05-17 AbbVie Inc. Novel tricyclic compounds
WO2011066980A2 (en) 2009-12-04 2011-06-09 Lars Holger Hermann Oral dosage forms with reduced potential for drug abuse
CA2784407A1 (en) 2009-12-17 2011-07-14 Cima Labs Inc. Abuse-resistant formulations
US8202525B2 (en) 2009-12-22 2012-06-19 Pondera Biotechnologies, LLC Methods and compositions for treating distress dysfunction and enhancing safety and efficacy of specific medications
US20110150989A1 (en) 2009-12-22 2011-06-23 Mallinkckrodt Inc. Methods of Producing Stabilized Solid Dosage Pharmaceutical Compositions Containing Morphinans
US8597681B2 (en) 2009-12-22 2013-12-03 Mallinckrodt Llc Methods of producing stabilized solid dosage pharmaceutical compositions containing morphinans
US9198861B2 (en) 2009-12-22 2015-12-01 Mallinckrodt Llc Methods of producing stabilized solid dosage pharmaceutical compositions containing morphinans
WO2011079074A1 (en) 2009-12-24 2011-06-30 Acura Phamaceuticals, Inc. Pharmaceutical compositions for deterring misuse, abuse, and diversion
US20130023553A1 (en) 2010-01-12 2013-01-24 Nektar Therapeutics Pegylated opioids with low potential for abuse and side effects
EP2359812A1 (en) 2010-01-18 2011-08-24 Cephalon France Oral lyophilised compositions
US20130022646A1 (en) 2010-02-09 2013-01-24 Rudnic Edward M Controlled Release Formulations of Opioids
US20110195989A1 (en) 2010-02-09 2011-08-11 Rudnic Edward M Controlled Release Formulations of Opioids
US20120321674A1 (en) 2011-02-17 2012-12-20 Michael Vachon Technology for Preventing Abuse of Solid Dosage Forms
US8501669B2 (en) 2010-02-10 2013-08-06 Companion Diagnostics Inc. Systems and methods of screening biomarkers in bodily fluids
US20110195520A1 (en) 2010-02-11 2011-08-11 Ameritox, Ltd. Methods of normalizing measured drug concentrations and testing for non-compliance with a drug treatment regimen
US9125867B2 (en) 2010-02-24 2015-09-08 Invincible Biotechnology Diversion- and/or abuse-resistant compositions and methods for making the same
WO2011107855A2 (en) 2010-03-04 2011-09-09 Torrent Pharmaceuticals Limited Sustained release oral liquid suspension dosage form
GB201003731D0 (en) 2010-03-05 2010-04-21 Univ Strathclyde Immediate/delayed drug delivery
GB201003766D0 (en) 2010-03-05 2010-04-21 Univ Strathclyde Pulsatile drug release
GB201003734D0 (en) 2010-03-05 2010-04-21 Univ Strathclyde Delayed prolonged drug delivery
US20110218209A1 (en) 2010-03-06 2011-09-08 Paul Daniel Yered Nutrient delievry drug composition
US20110230816A1 (en) 2010-03-18 2011-09-22 Tyco Healthcare Group Lp Gels for Transdermal Delivery
IT1398930B1 (en) 2010-03-24 2013-03-28 Molteni & C PHARMACEUTICAL FORMULATIONS BISTRATO CONTAINING OPPOSING AGONISTS AND ANTAGONISTS.
WO2011120084A1 (en) 2010-03-30 2011-10-06 Phosphagenics Limited Transdermal delivery patch
EP2552433A4 (en) 2010-03-30 2013-11-06 Algynomics Inc Compositions and methods for the treatment of somatosensory disorders
US20130224151A1 (en) 2010-03-31 2013-08-29 United States Of America Use of FAAH Inhibitors for Treating Abdominal, Visceral and Pelvic Pain
US20110239745A1 (en) 2010-04-01 2011-10-06 Lawrence Livermore National Security, Llc Rapid Identification of Explosives Using Thin-Layer Chromatography and Colorimetric Techniques
ES2592277T3 (en) 2010-04-02 2016-11-29 Buzzz Pharmaceuticals Limited Transdermal deterrent formulations of abuse of opioid agonists and agonists
WO2011127467A1 (en) 2010-04-09 2011-10-13 Companion Diagnostics, Inc. Devices, systems, and methods for biomarker stabilization
WO2011139595A2 (en) 2010-04-27 2011-11-10 Medtronic, Inc. Elongated biodegradable depot for sustained drug release to treat chronic pelvic pain
CA2798885C (en) 2010-05-10 2014-11-18 Euro-Celtique S.A. Combination of active loaded granules with additional actives
AU2011252040C1 (en) 2010-05-10 2015-04-02 Euro-Celtique S.A. Manufacturing of active-free granules and tablets comprising the same
FR2959935B1 (en) 2010-05-14 2013-02-08 Ethypharm Sa ALCOHOL-RESISTANT ORAL PHARMACEUTICAL FORM
CN101824144A (en) 2010-05-15 2010-09-08 西北师范大学 Polyethylene glycol aryloxyacetate, preparation thereof and application as plant growth regulator
CA3065589C (en) 2010-06-03 2022-04-26 Catalent Ontario Limited Multi phase soft gel capsules, apparatus and method thereof
JP2011256115A (en) 2010-06-04 2011-12-22 Michishi Tani Therapeutic drug for autism
FR2960775A1 (en) 2010-06-07 2011-12-09 Ethypharm Sa MICROGRANULES RESISTANT TO MISMATCH
AU2011271429B2 (en) 2010-06-30 2016-04-21 Upsher-Smith Laboratories, Llc Sustained release composition comprising an amine as active agent and a salt of a cyclic organic acid
US20120141554A1 (en) 2010-07-08 2012-06-07 Wellesley Pharmaceuticals, Llc Delayed release formulation for reducing the frequency of urination and method of use thereof
US20120015031A1 (en) 2010-07-14 2012-01-19 Grunenthal Gmbh Novel gastro-retentive dosage forms
CN101987081B (en) 2010-07-16 2012-08-08 钟术光 Controlled release preparation
US9526600B2 (en) 2010-07-20 2016-12-27 Warsaw Orthopedic, Inc. Biodegradable stents and methods for treating periodontal disease
US20120022009A1 (en) 2010-07-21 2012-01-26 Acella Pharmaceuticals, Llc Tannate dry powder formulations
CA2806362A1 (en) 2010-07-23 2012-01-26 Activbiotics Pharma, Llc Administration of rifalazil to immunocompromised patients
US20130303497A1 (en) 2010-08-05 2013-11-14 Conrig Pharma Aps Deuterated 5-ht1a receptor agonists
CH705678B1 (en) 2010-08-13 2016-03-15 Euro Celtique Sa Use of binders for the production of storage-stable formulations.
WO2012021819A1 (en) 2010-08-13 2012-02-16 Acura Pharmaceuticals, Inc. Optimized niacin compositions in pharmaceutical products
FR2963889B1 (en) 2010-08-20 2013-04-12 Debregeas Et Associes Pharma NALBUPHINE-BASED FORMULATIONS AND USES THEREOF
US8507844B2 (en) 2010-08-31 2013-08-13 Waters Technologies Corporation Techniques for sample analysis
CN101926757B (en) 2010-09-01 2013-01-02 北京大学 Liquid composition of indissolvable medicines and preparation method thereof
PE20131126A1 (en) 2010-09-02 2013-10-21 Gruenenthal Chemie ALTERATION RESISTANT DOSAGE FORM INCLUDING AN ANIONIC POLYMER
NZ607392A (en) 2010-09-02 2015-03-27 Gruenenthal Chemie Tamper resistant dosage form comprising inorganic salt
AR082861A1 (en) 2010-09-02 2013-01-16 Gruenenthal Gmbh ALTERATION RESISTANT DOSAGE FORM INCLUDING AN ANIONIC POLYMER
JP2013540748A (en) 2010-09-16 2013-11-07 コーネル ユニバーシティー Use of adenosine receptor signaling to regulate blood-brain barrier permeability
JP2013537915A (en) * 2010-09-24 2013-10-07 キューアールエックスファーマ リミテッド Opioid controlled release formulations
ES2933198T3 (en) 2010-10-21 2023-02-02 Rtu Pharmaceuticals Llc Ready-to-use ketorolac formulations
EP2444064A1 (en) 2010-10-22 2012-04-25 Meliatys Process for making multiparticulate gastroretentive dosage forms
US20120100183A1 (en) 2010-10-23 2012-04-26 Joel Schlessinger Topical base and active agent-containing compositions, and methods for improving and treating skin
US8685381B2 (en) 2010-10-23 2014-04-01 Joel Schlessinger Topical base and active agent-containing compositions, and methods for improving and treating skin
WO2012061779A1 (en) 2010-11-04 2012-05-10 Abbott Gmbh & Co. Kg Drug formulations
US20130230587A1 (en) 2010-11-10 2013-09-05 Rubicon Research Private Limited Sustained release compositions
EP2457900A1 (en) 2010-11-25 2012-05-30 Almirall, S.A. New pyrazole derivatives having CRTh2 antagonistic behaviour
CA2819663C (en) 2010-12-02 2019-12-24 Aptalis Pharmatech, Inc. Rapidly dispersing granules, orally disintegrating tablets and methods
EP2649443A2 (en) 2010-12-06 2013-10-16 Ramot at Tel-Aviv University Ltd Methods and kits for detection of drugs
GB201020895D0 (en) 2010-12-09 2011-01-26 Euro Celtique Sa Dosage form
US9233076B2 (en) 2010-12-13 2016-01-12 Purdue Pharma L.P. Controlled release dosage forms
ES2643291T3 (en) 2010-12-22 2017-11-22 Purdue Pharma L.P. Controlled release dosage forms with inviolable closure coated
CA2822769C (en) 2010-12-23 2016-10-04 Purdue Pharma L.P. Tamper resistant solid oral dosage forms
JP5903686B2 (en) 2010-12-23 2016-04-13 アッヴィ・ドイチュラント・ゲー・エム・ベー・ハー・ウント・コー・カー・ゲー Solid sustained release formulations based on solid dispersions
US8518438B2 (en) 2011-01-14 2013-08-27 Enspire Group, Llc Highly concentrated liquid acetaminophen solutions
WO2012098281A2 (en) 2011-01-19 2012-07-26 Universidad Miguel Hernández De Elche Trp-receptor-modulating peptides and uses thereof
USRE47009E1 (en) 2011-02-01 2018-08-28 The Children's Hospital Of Philadelphia HDAC inhibitors and therapeutic methods using the same
AU2012219322A1 (en) 2011-02-17 2013-05-09 QRxPharma Ltd. Technology for preventing abuse of solid dosage forms
JP6133790B2 (en) 2011-02-18 2017-05-24 ザ スクリプス リサーチ インスティテュート Directed differentiation of oligodendrocyte progenitor cells to myelinating cell fate
CN102648985B (en) 2011-02-24 2014-03-26 温州中科应急急救包有限公司 Chitosan emergent hemostasis material
AU2012222149B2 (en) 2011-02-25 2017-06-29 Arena Pharmaceuticals, Inc. Cannabinoid receptor modulators
WO2012116278A1 (en) 2011-02-25 2012-08-30 Arena Pharmaceuticals, Inc. Cannabinoid receptor modulators
KR101077468B1 (en) 2011-03-04 2011-11-07 (주)차바이오앤디오스텍 Stable orodispersible film formulation
CA2830882C (en) 2011-03-22 2021-03-16 Dinesh Barawkar Substituted fused tricyclic compounds, compositions and medicinal applications thereof
PL2688556T3 (en) 2011-03-25 2015-10-30 Purdue Pharma Lp Controlled release pharmaceutical dosage forms
BRPI1106121A2 (en) 2011-11-25 2015-12-08 Univ Minas Gerais pharmaceutical compositions containing 11,12-pyrazolimidocycline and use for neuropathic pain relief
TW201306825A (en) 2011-05-11 2013-02-16 Kirax Corp Package for improved treatment of conditions
US9050335B1 (en) 2011-05-17 2015-06-09 Mallinckrodt Llc Pharmaceutical compositions for extended release of oxycodone and acetaminophen resulting in a quick onset and prolonged period of analgesia
US8741885B1 (en) 2011-05-17 2014-06-03 Mallinckrodt Llc Gastric retentive extended release pharmaceutical compositions
EP2723452A2 (en) 2011-06-22 2014-04-30 Vyome Biosciences Pvt Ltd Conjugate-based antifungal and antibacterial prodrugs
US8623377B2 (en) 2011-06-29 2014-01-07 University Of Maryland, Baltimore Joint-homing peptides and uses thereof
HUE031251T2 (en) 2011-06-30 2017-07-28 Develco Pharma Schweiz Ag Controlled release oral dosage form comprising oxycodone
WO2013003845A1 (en) 2011-06-30 2013-01-03 Neos Therapeutics, Lp Abuse resistant drug forms
CN102344534A (en) 2011-07-12 2012-02-08 天津工业大学 Polymethyl polyglycol methacrylate containing paracetamol structure as well as preparation method and use method thereof
EP2548876A1 (en) 2011-07-18 2013-01-23 Almirall, S.A. New CRTh2 antagonists
EP2548863A1 (en) 2011-07-18 2013-01-23 Almirall, S.A. New CRTh2 antagonists.
US20130028955A1 (en) 2011-07-25 2013-01-31 Gaurav Thakersi Tolia Sustained release oral matrix and methods of making thereof
US8579924B2 (en) 2011-07-26 2013-11-12 Covidien Lp Implantable devices including a mesh and a pivotable film
KR101303479B1 (en) 2011-07-28 2013-09-06 (주)씨엘팜 Composition for edible film and pharmaceutical preparation for edible film containing drugs
EP2736495B1 (en) 2011-07-29 2017-08-23 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
DK2736497T3 (en) 2011-07-29 2017-11-13 Gruenenthal Gmbh Shock-resistant tablet that provides an immediate release of a drug.
CN102389423A (en) 2011-09-09 2012-03-28 北京阜康仁生物制药科技有限公司 Medicinal composition containing ibuprofen sodium salt
WO2013038268A1 (en) 2011-09-16 2013-03-21 Purdue Pharma L.P. Tamper resistant immediate release formulations
EP2755640B1 (en) 2011-09-16 2017-07-26 Purdue Pharma LP Tamper resistant pharmaceutical formulations
CA2847613A1 (en) 2011-09-16 2013-03-21 Purdue Pharma L.P. Tamper resistant immediate release formulations
AR088250A1 (en) 2011-10-06 2014-05-21 Gruenenthal Gmbh ORAL PHARMACEUTICAL DOSAGE FORM RESISTANT TO ALTERATION UNDERSTANDING OPIOID AGONIST AND OPIOID ANTAGONIST
EP2768537B1 (en) 2011-10-18 2019-02-20 Purdue Pharma LP Acrylic polymer formulations
EP2769219B1 (en) 2011-10-21 2017-08-30 Decimadx LLC Point-of care immunoassay for quantitative small analyte detection
WO2013058496A1 (en) 2011-10-21 2013-04-25 Daewoong Pharmaceutical Co., Ltd. Taste-masked pharmaceutical composition for oral administration and a process for the preparation thereof
KR101203186B1 (en) 2011-10-21 2012-11-23 주식회사 대웅제약 Taste-masked pharmaceutical composition for oral administration and a process for the preparation thereof
US8932621B2 (en) 2011-10-25 2015-01-13 Covidien Lp Implantable film/mesh composite
US9005308B2 (en) 2011-10-25 2015-04-14 Covidien Lp Implantable film/mesh composite for passage of tissue therebetween
US20130116215A1 (en) 2011-10-28 2013-05-09 Mireia Coma Combination therapies for treating neurological disorders
CA2854512A1 (en) 2011-11-07 2013-05-16 Nektar Therapeutics Compositions, dosage forms, and co-administration of an opioid agonist compound and an analgesic compound
US20130122098A1 (en) 2011-11-14 2013-05-16 Bayer Healthcare Llc Method and composition to improve absorption of therapeutic agents
WO2013072395A1 (en) 2011-11-17 2013-05-23 Grünenthal GmbH Tamper-resistant oral pharmaceutical dosage form comprising a pharmacologically active ingredient, an opioid antagonist and/or aversive agent, polyalkylene oxide and anionic polymer
US20150224097A1 (en) 2011-11-22 2015-08-13 Watson Pharmaceuticals, Inc. Immediate Release Abuse Deterrent Tablet
WO2013082308A1 (en) 2011-11-30 2013-06-06 Children's Hospital Medical Center Personalized pain management and anesthesia: preemptive risk identification and therapeutic decision support
FR2983409B1 (en) 2011-12-06 2013-12-27 Ethypharm Sa COMPRESSOR CAPABLE OF COMBATTING INJECTION MISTAKE
US20150265536A1 (en) 2011-12-09 2015-09-24 Purdue Pharma L.P. Pharmaceutical dosage forms comprising poly(epsilon-caprolactone) and polyethylene oxide
WO2013093877A2 (en) 2011-12-23 2013-06-27 Koninklijke Philips Electronics N.V. Encapsulation system for controlled release of a bleaching agent
US9095840B2 (en) 2012-01-03 2015-08-04 The Penn State Research Foundation Nitrogen-containing activated carbon material
EP2800776B1 (en) 2012-01-04 2020-04-29 Momentive Performance Materials Inc. Silicone adhesive compositions
GB201202223D0 (en) 2012-02-09 2012-03-28 Randox Lab Ltd Immunoassay for pyrrolidinophenones
WO2013119231A1 (en) 2012-02-09 2013-08-15 Tris Pharma, Inc. Abuse resistant opioid drug - ion exchange resin complexes having hybrid coatings
CN103040829B (en) 2012-02-17 2015-02-11 北京人福军威医药技术开发有限公司 Pharmaceutical composition containing lappaconitine and oxycodone
MX356421B (en) 2012-02-28 2018-05-29 Gruenenthal Gmbh Tamper-resistant dosage form comprising pharmacologically active compound and anionic polymer.
US20130225412A1 (en) 2012-02-28 2013-08-29 Soroush Sardari Lodriche Silicon nanocarrier for delivery of drug, pesticides and herbicides , and for waste water treatment
WO2013127830A1 (en) 2012-02-28 2013-09-06 Grünenthal GmbH Tamper-resistant pharmaceutical dosage form comprising nonionic surfactant
WO2013128447A1 (en) 2012-02-28 2013-09-06 Sipnose Ltd. Nasal delivery device
NZ629468A (en) 2012-03-02 2017-08-25 Rhodes Pharmaceuticals Lp Tamper resistant immediate release formulations
ES2637341T3 (en) 2012-03-14 2017-10-11 Levicept Ltd. P75NTR neurotrophin binding protein for therapeutic use
WO2013138118A1 (en) 2012-03-14 2013-09-19 The Regents Of The University Of California Treatment of inflammatory disorders in non-human mammals
EP2833883A4 (en) 2012-04-06 2015-12-23 Indus Pharmaceuticals Inc Novel compositions of combinations of non-covalent dna binding agents and anti-cancer and/or anti-inflammatory agents and their use in disease treatment
US9687445B2 (en) 2012-04-12 2017-06-27 Lts Lohmann Therapie-Systeme Ag Oral film containing opiate enteric-release beads
CA2868326A1 (en) 2012-04-13 2013-10-17 Banner Pharmacaps, Inc. Soft elastic capsules containing tablets and liquid or semisolid fills and methods for their manufacture
LT2838512T (en) 2012-04-18 2018-11-12 GrĆ¼nenthal GmbH Tamper resistant and dose-dumping resistant pharmaceutical dosage form
MX356111B (en) 2012-04-18 2018-05-15 SpecGx LLC Immediate release, abuse deterrent pharmaceutical compositions.
EP2838536B1 (en) 2012-04-18 2017-08-09 Mallinckrodt LLC Immediate release pharmaceutical compositions with abuse deterrent properties
EP2846755A1 (en) 2012-05-09 2015-03-18 SiO2 Medical Products, Inc. Saccharide protective coating for pharmaceutical package
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
WO2013171146A1 (en) 2012-05-15 2013-11-21 Lts Lohmann Therapie-Systeme Ag Oral film containing enteric release opiate resinate
WO2013175511A1 (en) 2012-05-24 2013-11-28 V.B. Medicare Pvt. Ltd. Taste masked pharmaceutical compositions
JP2013249458A (en) 2012-06-04 2013-12-12 Hitachi Chemical Co Ltd Epoxy resin molding material for sealing and electronic component device
DE102012105512A1 (en) 2012-06-25 2014-04-24 Hennig Arzneimittel Gmbh & Co. Kg Pharmaceutical form for prolonged release of active ingredients
WO2014004126A1 (en) 2012-06-26 2014-01-03 Fleming C Andrew Treating postoperative nausea and vomiting
ES2938586T3 (en) 2012-06-27 2023-04-12 Medincell S A Biodegradable drug release for hydrophobic compositions
EP2872121B1 (en) 2012-07-12 2018-09-05 SpecGx LLC Extended release, abuse deterrent pharmaceutical compositions
US20140045801A1 (en) 2012-08-09 2014-02-13 Mylan Inc. Pramipexole transdermal delivery for severe headaches
WO2014045305A1 (en) 2012-09-21 2014-03-27 Advinus Therapeutics Limited Substituted fused tricyclic compounds, compositions and medicinal applications thereof
CA2791206A1 (en) 2012-09-28 2014-03-28 Pharmascience Inc. Abuse deterrent pharmaceutical formulation
US10987365B2 (en) 2012-10-01 2021-04-27 Gm Pharmaceuticals, Inc. Compositions and methods for the treatment of pain
WO2014059512A1 (en) 2012-10-15 2014-04-24 Isa Odidi Oral drug delivery formulations
CN103070840A (en) 2012-10-31 2013-05-01 北京正大绿洲医药科技有限公司 Oxycodone hydrochloride slow release dripping pills and preparation method thereof
CN113712911A (en) * 2013-03-15 2021-11-30 维普詹尼克斯公司 Novel analgesic composition
WO2015023704A1 (en) 2013-08-12 2015-02-19 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
CA3042642A1 (en) 2013-08-12 2015-02-19 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
CN103637987B (en) 2013-12-09 2015-12-02 韩彬 The composition of liquid medicine of oxycodone
CN103637998B (en) 2013-12-09 2016-04-06 韩彬 Comprise the slow releasing tablet of oxycodone and rotundine
US10772841B2 (en) 2014-04-07 2020-09-15 Patheon Softgels Inc. Opioid abuse-deterrent controlled release formulations

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4450877A (en) * 1977-11-03 1984-05-29 Hoechst Aktiengesellschaft Pharmaceutical preparations in solid unit dosage form
WO1991007950A1 (en) * 1989-11-24 1991-06-13 Farmitalia Carlo Erba Limited Pharmaceutical compositions
US20090123386A1 (en) 2005-04-06 2009-05-14 Victor Morrison Young Abuse Resistant Capsules
WO2008021394A2 (en) * 2006-08-15 2008-02-21 Theraquest Biosciences, Llc Pharmaceutical formulations of cannabinoids and method of use
US20100204259A1 (en) 2009-02-06 2010-08-12 Egalet A/S Immediate release composition resistant to abuse by intake of alcohol
US20140171481A1 (en) * 2010-06-10 2014-06-19 Abbvie, Inc. Solid compositions
US20140010873A1 (en) 2012-07-06 2014-01-09 Egalet Ltd. Abuse deterrent pharmaceutical compositions for controlled release

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"Guidance for Industry: Abuse -Deterrent Opioids - Evaluation and Labeling", January 2013, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, FDA, CDER

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3209282A4 (en) * 2014-10-20 2018-05-23 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form

Also Published As

Publication number Publication date
AU2019200026A1 (en) 2019-01-24
EP3169315A4 (en) 2018-03-21
ES2809458T3 (en) 2021-03-04
JP2017521448A (en) 2017-08-03
EP3169315B1 (en) 2020-06-24
EP3169315A1 (en) 2017-05-24
AU2015290098B2 (en) 2018-11-01
AU2015290098A1 (en) 2017-01-19
JP6371463B2 (en) 2018-08-08
US9707184B2 (en) 2017-07-18
DK3169315T3 (en) 2020-08-10
CA2955229A1 (en) 2016-01-21
US20170340574A1 (en) 2017-11-30
CA2955229C (en) 2020-03-10
US20160015650A1 (en) 2016-01-21
JP2018197241A (en) 2018-12-13

Similar Documents

Publication Publication Date Title
CA2913368C (en) Abuse deterrent immediate release formulation
CA2919892C (en) Extruded immediate release abuse deterrent pill
TWI463983B (en) Tamper resistant dosage forms
EP3473246A1 (en) Immediate release abuse deterrent formulations
US9707184B2 (en) Immediate release abuse deterrent liquid fill dosage form
US10959958B2 (en) Extended release abuse deterrent liquid fill dosage form
CA3002181C (en) Food independent immediate release drug formulation with abuse deterrence and overdose protection
AU2019216710A1 (en) Extruded extended release abuse deterrent pill
EP3032948A2 (en) Extruded immediate release abuse deterrent pill
WO2014197137A1 (en) Diversion-resistant formulation

Legal Events

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

Ref document number: 15821947

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2955229

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017502652

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015821947

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015821947

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015290098

Country of ref document: AU

Date of ref document: 20150707

Kind code of ref document: A