WO2017003935A1 - Ketamine transdermal delivery system - Google Patents

Ketamine transdermal delivery system Download PDF

Info

Publication number
WO2017003935A1
WO2017003935A1 PCT/US2016/039601 US2016039601W WO2017003935A1 WO 2017003935 A1 WO2017003935 A1 WO 2017003935A1 US 2016039601 W US2016039601 W US 2016039601W WO 2017003935 A1 WO2017003935 A1 WO 2017003935A1
Authority
WO
WIPO (PCT)
Prior art keywords
transdermal delivery
ketamine
delivery device
provides
present
Prior art date
Application number
PCT/US2016/039601
Other languages
French (fr)
Inventor
Huadong Tang
Hock S. Tan
Michael MAYERSOHN
Original Assignee
Shenox Pharmaceuticals, Llc
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 Shenox Pharmaceuticals, Llc filed Critical Shenox Pharmaceuticals, Llc
Priority to JP2017566151A priority Critical patent/JP6845162B2/en
Priority to KR1020187002315A priority patent/KR102299359B1/en
Priority to AU2016288188A priority patent/AU2016288188B2/en
Priority to CN201680037543.8A priority patent/CN107847469A/en
Priority to US15/736,182 priority patent/US11191734B2/en
Priority to EP16818548.6A priority patent/EP3297620A4/en
Priority to CA2987909A priority patent/CA2987909C/en
Publication of WO2017003935A1 publication Critical patent/WO2017003935A1/en
Priority to HK18108421.2A priority patent/HK1248578A1/en
Priority to US17/540,695 priority patent/US20220193000A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • A61K9/703Transdermal patches and similar drug-containing composite devices, e.g. cataplasms characterised by shape or structure; Details concerning release liner or backing; Refillable patches; User-activated patches
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • A61K9/703Transdermal patches and similar drug-containing composite devices, e.g. cataplasms characterised by shape or structure; Details concerning release liner or backing; Refillable patches; User-activated patches
    • A61K9/7038Transdermal patches of the drug-in-adhesive type, i.e. comprising drug in the skin-adhesive layer
    • A61K9/7046Transdermal patches of the drug-in-adhesive type, i.e. comprising drug in the skin-adhesive layer the adhesive comprising macromolecular compounds
    • A61K9/7053Transdermal patches of the drug-in-adhesive type, i.e. comprising drug in the skin-adhesive layer the adhesive comprising macromolecular compounds obtained by reactions only involving carbon to carbon unsaturated bonds, e.g. polyvinyl, polyisobutylene, polystyrene
    • A61K9/7061Polyacrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • A61K9/703Transdermal patches and similar drug-containing composite devices, e.g. cataplasms characterised by shape or structure; Details concerning release liner or backing; Refillable patches; User-activated patches
    • A61K9/7038Transdermal patches of the drug-in-adhesive type, i.e. comprising drug in the skin-adhesive layer
    • A61K9/7046Transdermal patches of the drug-in-adhesive type, i.e. comprising drug in the skin-adhesive layer the adhesive comprising macromolecular compounds
    • A61K9/7053Transdermal patches of the drug-in-adhesive type, i.e. comprising drug in the skin-adhesive layer the adhesive comprising macromolecular compounds obtained by reactions only involving carbon to carbon unsaturated bonds, e.g. polyvinyl, polyisobutylene, polystyrene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants

Definitions

  • the present invention is directed to transdermal delivery devices comprising ketamine and formulations thereof.
  • the present invention is also directed to transdermal delivery devices comprising ketamine for the treatment of major depressive disorder (MDD) and/or pain.
  • MDD major depressive disorder
  • the present invention is further directed to transdermal delivery devices comprising ketamine and abuse deterrent agents.
  • MDD Major depressive disorder
  • MAOIs monoamine oxidase inhibitors
  • SNRIs serotonin-norepinephrine reuptake inhibitors
  • SSRIs selective serotonin reuptake inhibitors
  • the delay of onset varies from weeks to months, which may result in adverse events, including but not limited to increased vulnerability to suicide, decrease in compliance, and increase in social and economic burden.
  • Common side effects of these antidepressants include nausea, insomnia, anxiety, weight- loss/gain, drowsiness, headache, loss of sex drive, and/or blurred vision.
  • One type of pain, neuropathic pain is a complex chronic pain state often accompanied by tissue injury. The occurrence of pain with neuropathic characteristics is about 6.9-10% of the general population. Hecke et al , Pain, 155(4):654-62 (2014). Symptoms of neuropathic pain include spontaneous burning, shooting pain, hyperalgesia, and allodynia. Patients with neuropathic pain often have conditions that are associated with other significant health issues, including depression, sleep problems, and loss of independence. Bouhassira ei a/., Pain., 136(3):380-7 (2008).
  • Neuropathic pain can be caused by a variety of mechanisms, including infection, central or peripheral nerve injury, stroke, multiple sclerosis, diabetes mellitus, sarcoidosis, toxic agents (e.g., alcohol or chemotherapy), inherited or genetic neuropathy, and Complex Regional Pain Syndrome (CRPS).
  • CRPS is an intractable form of pain, often resistant to a variety of conventional therapies.
  • Correll et al Pain Med., 5(3):263-75 (2004).
  • Pain Med., 5(3):263-75 (2004) Pain Med., 5(3):263-75 (2004).
  • Treatment for neuropathic pain includes antidepressants, anticonvulsants, and/or topical pain management medications.
  • Niesters et al Expert Opin. Drug Metab. Toxicol, 8(11): 1409-17 (2012); Dworkin ei al, Pain, 132(3):237-51 (2007).
  • Ketamine is a non-competitive, N-methyl-D-aspartate (NMDA) receptor antagonist, indicated for treatment as an anesthetic, sedative, and analgesic. Ketamine has been demonstrated to be an effective antidepressant, with rapid onset (within about 2 hours of administration) and sustained antidepressant effect (from days to in some cases a week or two after administration). Berman et al, Biol. Psychiatry, 47(4):351-54 (2000). The NMDA receptor pathway plays an important role in pain, including neuropathic pain. Animal studies and human clinical studies have shown the efficacy of ketamine in the treatment of chronic neuropathic pain. Correll et al , Pain Med. 5(3):263-75 (2004); Sigtermans et al, Pain, 145(3):304-11 (2009).
  • NMDA N-methyl-D-aspartate
  • Ketamine is a racemic mixture containing R-ketamine and S-ketamine. It is generally believed that the anesthetic and/or antidepressant effect of ketamine is mainly through the action of S-ketamine because in vitro S-ketamine has about a 4-fold greater affinity than the R-ketamine on NMDA receptor binding. However, animal model studies have suggested that R-ketamine is more effective as an antidepressant than S-ketamine. In addition, R-ketamine was shown to be free of psychotomimetic side effects and abuse liability. Yang et al , Transl. Psychiatry, 5(e632): l-l l (2015). The present invention is directed toward administration of the racemic mixture of ketamine; however, embodiments containing the R-ketamine or the S- ketamine enantiomers are within the scope of the present invention.
  • Ketamine is also a known dissociative anesthetic that has gained popularity as a drug of abuse, and may be referred to illicitly as "K” or "Special K". Ketamine is reported to distort perceptions of sight and sound, and make the user feel disconnected.
  • MTF Monitoring the Future
  • Illicit ketamine can be distributed as a dried powder or as a liquid, mixed with beverages, and/or added to smokable materials (such as marij uana or tobacco). As a powder, ketamine can be snorted or pressed into tablets, sometimes in combination with other drugs, including 3,4-methylenedioxymethamphetamine (MDMA, referred to illicitly as "ecstasy”), amphetamine, methamphetamine, cocaine, and/or carisoprodol.
  • MDMA 3,4-methylenedioxymethamphetamine
  • ecstasy 3,4-methylenedioxymethamphetamine
  • amphetamine methamphetamine
  • cocaine carisoprodol
  • IV administration of ketamine presents numerous challenges.
  • Cmax maximum plasma concentration
  • ketamine has a short half-life (about 2 hours)
  • this immediate release delivery of ketamine by IV administration may result in little to no ketamine remaining in plasma after about 4-8 hours, necessitating frequent and repeated dosing to maintain therapeutic plasma levels.
  • intranasal formulation of the S-enantiomer of ketamine, esketamine is under development and in clinical study by Janssen. US 2013/0236573 Al, Singh et al, Esketamine For The Treatment of Treatment-Refractory Or Treatment-Resistant Depression.
  • intranasal delivery of ketamine presents numerous challenges. It suffers from many of the same immediate release issues faced by IV administration of ketamine, namely, rapid onset of maximum concentration (Tmax), high Cmax, increased risk of side effects like drug toxicity, and the need for frequent and multiple dosing to maintain therapeutic plasma concentrations. Frequent administration of intranasal ketamine may increase the risk of irritating and damaging the nasal epithelium, which in turn may reduce patient compliance.
  • intranasal administration is associated with high variability in absorption among subjects. Kublik et al, Adv. Drug Deliv. Rev. 29: 157-77 (1998). Further, the rapid rise in ketamine plasma concentration following intranasal administration may cause adverse side effects, such as drug toxicity. Moreover, intranasal delivery of ketamine, without additional safeguards, is highly susceptible to abuse. Other routes of administration of ketamine, including parenteral administration of ketamine (e.g., subcutaneous, intramuscular, etc.) suffer from many of these same challenges.
  • Ketamine has a high systemic (primarily hepatic) clearance of about 19 ml/min»kg, a rate which approaches liver plasma flow.
  • ketamine is subject to substantial pre- systemic metabolism, or first-pass effect, in the liver and gut wall by metabolic enzymes, such as cytochrome P450 enzymes (CYP450). Consequently, the absolute oral bioavailability of ketamine in humans is only about 10-20%. Due to this first-pass effect, there is an increased risk for drug-drug interactions (DDI) with drugs that can inhibit or induce CYP450s. Clements et al.
  • the present invention is directed to transdermal delivery devices comprising ketamine and formulations thereof.
  • the present invention is also directed to transdermal delivery devices comprising ketamine for the treatment of major depressive disorder (MDD) and/or pain.
  • MDD major depressive disorder
  • MDD major depressive disorder
  • the controlled, prolonged, and steady ketamine exposure to humans from the transdermal delivery device of the present invention can reduce adverse side effects compared with other routes of ketamine delivery, including but not limited to intravenous (IV) administration and intranasal spray.
  • IV intravenous
  • the present invention is further directed to transdermal delivery devices comprising ketamine and abuse deterrent agents.
  • the present invention has numerous advantages. Formulations of the transdermal delivery device provide excellent ketamine permeability and stability. The inventors discovered, through in vitro experiments, that ketamine has excellent transdermal permeability properties, which are very important for efficacious clinical use. Additional in vitro experiments demonstrated that adding, for example, crystallization inhibitors to formulations of the present invention resulted in very stable transdermal delivery devices, which is important when making a pharmaceutical product.
  • the present invention provides improved drug metabolism and pharmacokinetic properties compared with other methods of administering ketamine, such as IV and intranasal administration.
  • transdermal delivery avoids the aforementioned first- pass effect.
  • Correll et al Pain Med. 5(3):263-75 (Sept. 2004). Instead, the present invention can meet the desired prolonged drug absorption profile.
  • administration of a single transdermal delivery device of the present invention can deliver a relatively constant ketamine plasma concentration for up to about 7 days.
  • the present invention delivers lower Cmax values for ketamine in the plasma, as well as minimal fluctuation between Cmax and Cmin, thereby reducing adverse side effects, including but not limited to toxicity, psychotropic effects, increased potential for addiction, and lack of therapeutic effect.
  • the transdermal delivery device of the present invention provides flexibility in dose, dosage release rate, patch size, and duration of application to allow for optimization.
  • Transdermal delivery devices according to the present invention include, but are not limited to, transdermals and dermal patches, topical skin applications such as spray, creams, gels, lotions, dressings and liquid solutions, and other transdermal delivery systems and dosage forms known to persons skilled in the art.
  • these flexible parameters can be adjusted by the formulator and/or the clinician to provide the optimal ketamine plasma concentration-time profile for the individual patient that maximizes efficacy and minimizes adverse side effects. Therefore, the ketamine transdermal delivery device of the present invention is particularly effective for the treatment of MDD and pain.
  • the present invention improves convenience and compliance compared with other forms of administering ketamine.
  • administration of the transdermal delivery device once or twice a week is more convenient than, for example, multiple daily doses of immediate release forms of ketamine.
  • Immediate release ketamine or immediate release ketamine formulation means administration of ketamine that is not extended, controlled, delayed or prolonged. Dose equivalent with means that the total dosage of drug administered between the compared items is the same.
  • the present invention is less invasive and less costly than IV administration. The present invention causes less irritation and is less invasive than intranasal administration. Further, the present invention is less likely to cause drug toxicity than immediate release forms of drug delivery.
  • the present invention also has advantages with respect to abuse deterrence.
  • the transdermal delivery device itself may serve as an abuse deterrent because it is more difficult to abuse directly, such as by biting or swallowing the device.
  • the ketamine is incorporated into a polymeric matrix together with other excipients, including, but not limited to skin permeation enhancers, humectants, plasticizers, buffers, antioxidants, and combinations thereof, each of which may inhibit ketamine extraction for abuse. Nevertheless, specific, additional abuse deterrent agents can be added to formulations of the present invention to further deter abuse.
  • Figure 1 is a ketamine transdermal delivery device without abuse deterrent properties.
  • Figure 2 is a transdermal delivery device comprising ketamine with abuse deterrent properties.
  • Figure 3 is an alternate embodiment of a transdermal delivery device comprising ketamine and abuse deterrent agents.
  • Figure 4 is an alternate embodiment of a transdermal delivery device comprising ketamine and abuse deterrent agents.
  • Figures 5-7 are ketamine plasma concentration versus time profiles in humans for Example 2 of the present invention using different sizes of the transdermal delivery device. These pharmacokinetic profiles are predicted by known convolution methodology using in vitro transdermal permeation data and in vivo intravenous plasma concentration data.
  • Figure 8 is a graph of the in vitro skin permeation of ketamine in a Franz Diffusion Cell model for the transdermal delivery device according to Example 2.
  • Figure 9 is a 6-month stability graph of the present invention prepared in accordance with Example 5.
  • Figures 10-13 are ketamine plasma concentration versus time profiles in humans corresponding to the transdermal delivery devices of Table 2. These transdermal delivery devices are for the treatment of MDD. These pharmacokinetic profiles are predicted by known convolution methodology using in vitro transdermal permeation data and in vivo intravenous plasma concentration data.
  • Figures 14-17 are graphs of the pharmacokinetic plasma concentrations versus time profiles, corresponding to the transdermal delivery devices of Table 3. These transdermal delivery devices are for the treatment of pain. These pharmacokinetic profiles are predicted by known convolution methodology using in vitro transdermal permeation data and in vivo intravenous plasma concentration data.
  • the active ingredient in the transdermal delivery device of the present invention is preferably employed at about 1-35% by weight of the final formulation (also referred to as 1- 35 weight percent) and most preferably 10-25% by weight of the final formulation.
  • the most preferred active ingredient is ketamine.
  • Additional active ingredients that can be employed in the present invention can be chosen from drugs that counteract the potential adverse effects of ketamine, reduce ketamine addiction potential, and/or enhance ketamine's antidepressant effect and/or pain management effects. All weight percentages provided in the present disclosure are based on the weight of the final formulation, which includes the adhesive-drug layer (or adhesive-drug matrix) and the abuse deterrent layer (or abuse deterrent matrix, which is optional), but not the release liner or the backing film.
  • the transdermal permeation rate (mg/day) combined with the size of the transdermal delivery device and the duration of application of the transdermal delivery device determines the plasma concentration of the drug.
  • the transdermal permeation rate of the ketamine transdermal delivery device of the present invention will preferably be about 0.1-30 mg/day/cm 2 of the transdermal delivery device, and most preferably about 0.5-5 mg/day/cm 2 .
  • the size of transdermal delivery device will preferably be about 5 -300 cm 2 .
  • the duration of application of the transdermal devices will preferably be about 8-168 hours. Combinations of these preferred ranges provide prolonged plasma concentrations of ketamine ranging from about 0.4-3850 ng/ml.
  • transdermal permeation rates a preferred transdermal permeation rate for the transdermal delivery devices of the present invention.
  • Plasma concentrations provided by any given dosage strength for the transdermal delivery device of the present invention can vary from individual to individual.
  • the preferred plasma concentration range of ketamine is from about 10-200 ng/ml, and the most preferred plasma concentration range is from about 10-100 ng/ml.
  • Table 2 provides ranges for the most preferred transdermal permeation rates, transdermal delivery device sizes, and duration of application of the transdermal delivery devices for antidepressant effect (i.e. , the treatment of MDD) with minimized adverse side effects. These ranges are chosen to provide prolonged plasma concentrations of ketamine ranging from about 10 -200 ng/ml for about 8-168 hours, and the most preferred plasma concentrations are from about 10-100 ng/ml.
  • Variations on the transdermal delivery devices comprising ketamine designed for the treatment of MDD will contain from about 8.3-200 mg and be applied for about 8 hours, from about 25-600 mg and be applied for about 24 hours, from about 87.5-2100 mg and be applied for about 84 hours, and from about 175-4200 mg and be applied for about 168 hours.
  • Transdermal delivery devices for the treatment of MDD are prepared with dosage strengths from about 8.3-200 mg, and are designed to be applied for about 8 hours, which will provide a plasma concentration of ketamine from about 11-257 ng/ml.
  • An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects.
  • the reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 8.3-100 mg of ketamine, designed to be applied for 8 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
  • Transdermal delivery devices for the treatment of MDD according to the present invention are prepared with dosage strengths from about 25-600 mg and are designed to be applied for about 24 hours, which will provide a plasma concentration of ketamine from about 11 -257 ng/ml.
  • An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects.
  • the reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 25-300 mg of ketamine, designed to be applied for 24 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
  • Transdermal delivery devices for the treatment of MDD according to the present invention are prepared with dosage strengths from about 87.5-2100 mg, and are designed to be applied for about 84 hours, which will provide a plasma concentration of ketamine from about 11 -257 ng/ml.
  • An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects.
  • the reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 87.5-1050 mg, designed to be applied for about 84 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
  • Transdermal delivery devices for the treatment of MDD according to the present invention are prepared with dosage strengths from about 175-4200 mg, and are designed to be applied for about 168 hours, which will provide a plasma concentration of ketamine from about 11 -257 ng/ml.
  • An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects.
  • the reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 175-2100 mg, designed to be applied for about 168 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
  • the preferred plasma concentrations ranges of ketamine are from about 50-1000 ng/ml, and the most preferred plasma concentration is about 500 ng/ml.
  • Table 3 provides ranges for transdermal permeation rates, transdermal delivery device sizes, and duration of application of the transdermal delivery devices for pain management. These ranges are chosen to provide prolonged plasma concentrations of ketamine ranging from about 50-1000 ng/ml, and for about 8-168 hour, and the most preferred plasma concentration is about 500 ng/ml.
  • Transdermal delivery devices comprising ketamine designed for pain management will contain about 40-500 mg and be applied for about 8 hours, from about 120- 1500 mg and be applied for about 24 hours, from about 420-5250 mg and be applied for about 84 hours, and from about 840-10500 mg and be applied for about 168 hours.
  • Transdermal delivery devices for the treatment of pain according to the present invention are prepared with dosage strengths from about 40-500 mg, are designed to be applied for about 8 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
  • Transdermal delivery devices for the treatment of pain according to the present invention are prepared with dosage strengths from about 120-1500 mg, are designed to be applied for about 24 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
  • Transdermal delivery devices for the treatment of pain are prepared with dosage strengths from about 420-5250 mg, are designed to be applied for about 84 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
  • Transdermal delivery devices for the treatment of pain are prepared with dosage strengths from about 840-10,500 mg, are designed to be applied for about 168 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
  • the transdermal delivery device of the present invention will be administered once a day, twice a week, or once a week.
  • the dosing regimen of the present invention is not limited to the examples provided in Tables 2 and 3 for antidepressant effect and pain management.
  • the dose frequencies, device size, and/or dosage strength can be adjusted.
  • the application of the transdermal delivery device can be for a duration shorter than 8 hours, such as 4 hours.
  • the ketamine plasma concentrations at about 4 hours after administration will be about 80% of the plasma concentration at about 8 hours, which can provide effective anti-depressant and/or pain management, depending on the need of the patient.
  • 6-7 are exemplary of the plasma profiles of the present invention.
  • the plasma drug concentration profiles in Tables 2 and 3 and illustrated at least in 10-17, are further exemplary of the plasma profiles of the present invention. These plasma profiles rise slowly and are maintained at a relatively constant level for a prolonged period of time.
  • IV and intranasal ketamine will generally provide a Cmax approximately 3 to 10 times higher than the Cmax provided by a transdermal delivery device of the present invention (at equivalent doses), while the area under the curve (AUC) is constant (e.g. , Figure 5).
  • the prolonged and steady administration of ketamine provided by the transdermal delivery device of the present invention exhibits minimal fluctuations in plasma concentration relative to multiple doses of IV or intranasal administration of ketamine. This reduction or minimization of plasma fluctuations in turn reduces the occurrence of adverse side effects resulting from under and over medication. Consequently, the plasma profile provided by the present invention is improved and may result in better therapeutic outcomes and greater patient compliance.
  • the structure and packaging of the transdermal delivery device of the present invention are prepared in accordance with methods and techniques known to persons skilled in the art.
  • the primary components are the backing layer, the adhesive-drug layer (or adhesive-drug matrix), the abuse deterrent layer (or abuse deterrent matrix) (optional), and the release liner.
  • the backing layer may be comprised of polymeric films such as polyester (PET) or polyethylene (PE) films that support the adhesive drug matrix and protect the transdermal delivery device from the environment.
  • PET polyester
  • PE polyethylene
  • the preferred thickness range for the backing film is from about 2-5 mils (1 mil equals 1/1000 of an inch), and the most preferred thickness range of the backing layer is from about 3-4 mils thick.
  • the adhesive in the adhesive-drug layer may be a pressure sensitive adhesive (PSA). Tan et al , Pharm Sci. & Tech Today , 2:60-69 (1999).
  • PSAs in transdermal delivery systems include, but are not limited to, polyisobutylenes (PIB), silicone polymers, and acrylate copolymers, such as acrylic pressure sensitive adhesives, including Duro-Tak 87-2516, 87- 2852 and 87-2194, manufactured by Henkel Adhesives.
  • PIBs are elastomeric polymers that are commonly used in PSAs, both as primary-base polymers and as tackifiers.
  • PIBs are homopolymers of isobutylene and feature a regular structure of a carbon-hydrogen backbone with only terminal unsaturation. PIBs are marketed under the trade name Oppanol by BASF.
  • the silicone polymers are a high molecular weight polydimethylsiloxane that contains residual silanol functionality (SiOH) on the ends of the polymer chains.
  • Silicone PSAs for use in pharmaceutical applications are available from Dow Corning Corporation, for example under the trade name of BIO-PSA.
  • the PSA is preferably employed at about 30-90% by weight of the final formulation, and most preferably about 40-60% by weight of the final formulation.
  • the release liner can be manufactured in the desired size for the present invention.
  • the release liner may be comprised of silicone or fluoro-polymer coated polyester film.
  • the release liner protects the transdermal delivery device during storage and is removed before its use.
  • Silicone-coated release liners are manufactured by Mylan Corporation, Loparex Corporation, and 3M's Drug Delivery Systems.
  • the fluoro-polymer coated release liners are manufactured and supplied by 3M's Drug Delivery Systems and Loparex.
  • the preferred thickness of the release liner is about 2-10 mils, and most preferably about 3-5 mils.
  • Additional drugs can be incorporated in the transdermal delivery device to counteract adverse effects, and/or to enhance the antidepressant or pain management effect of ketamine.
  • Examples for enhancing antidepressant effect include, but are not limited to, antagonists of group II metabotropic glutamate receptors, such as LY341495, Podkowa et al., Psychopharmacology (Berl) (Jun 11 2016).
  • Examples for reducing side effects with ketamine, especially psychotomimetic and sympathomimetic include, but not limited to, coadministration of alpha-2 agonists such as clonidine. Lenze, World J Biol Psychiatry, 17(3):230-8 (2016).
  • an additional drug is employed in the present invention, it is preferably employed at about 0.1-20% by weight of the final formulation, and most preferably about 1- 5% by weight of the final formulation.
  • Skin permeation enhancers are employed to enhance the skin permeability of the drug through the skin.
  • Skin permeation enhancers that may be employed in the present invention include, but are not limited to, sulphoxides (e.g. dimethylsulphoxide, DMSO), Azones (e.g. laurocapram), pyrrolidones (e.g. 2-pyrrolidone, 2P), alcohols and alkanols (ethanol, or decanol), glycols (e.g. propylene glycol (PG)), surfactants and terpenes. Williams et al. , Adv Drug Deliv Rev. 27;56(5):603-l 8 (2004).
  • the skin permeation enhancers are preferably employed at about 1-20% by weight of the final formulation, and most preferably about 4-10% by weight of the final formulation.
  • Humectants are employed to keep the transdermal delivery device hydrated and/or to reduce the loss of moisture.
  • the humectants that may be employed in the present invention include, but are not limited to, propylene glycol, glycerol, urea, polyvinylpyrrolidone (PVP), vinylpyrrolidone-vinyl acetate copolymers, and copolymers of PVP (e.g., BASF's Kollidon K30, K12, Kollidon VA 64, or Kollidon CL-M, magnesium silicate, and silica.
  • PVP polyvinylpyrrolidone
  • the humectants are preferably employed at about 2-20% by weight of the final formulation and most preferably about 5-10% by weight of the final formulation.
  • Plasticizers are employed in transdermal drug delivery systems to obtain desirable mechanical properties, such as to improve the film forming properties and the appearance of the film, to decrease the glass transition temperature of the polymer, to prevent film cracking, and to increase film flexibility.
  • the plasticizers that may be employed in the present invention include, but are not limited to, phthalate esters, phosphate esters, fatty acid esters, and glycol derivatives.
  • the plasticizers are preferably employed at about 2-20% by weight of the final formulation and most preferably about 5-10% by weight of the final formulation. Designing and Characterization of Drug Free Transdermal delivery devices for Transdermal Application, International Journal of Pharmaceutical Sciences and Drug Research, Vol. 2, No. 1 , pp.
  • Antioxidants are employed to prevent drug degradation by oxidation.
  • Antioxidants that may be employed in the present invention include, but are not limited to, butylated hydroxyanisole (BHA), butylhydroxy toluene (BHT), tert-Butylhydroquinone, ascorbic acid, and tocopherols.
  • BHA butylated hydroxyanisole
  • BHT butylhydroxy toluene
  • tert-Butylhydroquinone ascorbic acid
  • tocopherols tocopherols.
  • the antioxidants are preferably employed at about 0.01 -5% by weight of the final formulation and most preferably about 0.1-1.0% by weight of the final formulation.
  • Anti-irritants are employed to provide alleviation or prevention of skin irritation, and to assist in the release of the active ingredients.
  • Anti-irritants that may be employed in the present invention include, but are not limited to, aloe, arnica, chamomile, cucumber, menthol, mugwort, oat, zinc oxide, drug release modifiers such as chitosan, cellulose-based polymers, silicon dioxides, and polymethacrylates.
  • FIG. 1 is an embodiment of the transdermal delivery device in which the backing film
  • the adhesive drug matrix contains the drug and adhesive, as well as enhancers, humectants, plasticizers, antioxidants, pH modifiers, crystallization inhibitors, and other ingredients that aid in drug release and permeation through skin, and in maintaining drug stability.
  • Figure 2 is an example of a transdermal delivery device that contains an abuse deterrent agent (4) that is not skin permeable, drug (5) dissolved in the adhesive-drug matrix (2), a transdermal delivery device backing film (1), and a release liner (3).
  • Figure 3 is a transdermal delivery device that contains a backing film (1), an abuse deterrent layer (6), an adhesive-drug matrix (2), and a release liner (3).
  • the abuse deterrent layer is capable of releasing an abuse deterrent agent upon tampering with the transdermal delivery device.
  • the abuse deterrent layer comprises a gel forming agent
  • the gel forming agent can form a gel solution upon extraction.
  • Figure 4 is an embodiment of a prolonged use, e.g., 7-day transdermal delivery device, with abuse deterrent agents in a segregated, abuse deterrent layer.
  • Figure 4 shows the backing film (1), and overlay adhesive layer (7), an abuse deterrent layer (6), the adhesive-drug layer
  • the overlay adhesive layer extends over the outer edges of the drug-adhesive layer (2) and the abuse deterrent layer (6) to provide added adhesion to the skin for prolonged use.
  • Embodiments of the present invention can be prepared with an overlay adhesive layer (7) with or without an intervening abuse deterrent layer (6).
  • Table 4 provides estimated plasma concentrations for transdermal delivery devices prepared according to the present invention.
  • the plasma concentration are exemplified in Figs. 5, 6 and 7, which are described below in detail.
  • Table 5 provides the cumulative amount of ketamine that permeates human skin in the Franz Diffusion Cell model disclosed in Figure 8, as provided by a transdermal delivery device according to Example 2 of the present invention.
  • the total amount of drug in the transdermal delivery device of Example 2 is 4.75 mg. Therefore, the transdermal bioavailability of Example 2 within 24 hours is about 78%.
  • Transdermal delivery devices can be abused.
  • One method of abuse is to place the device in a solvent to separate the drug from the polymeric matrix, followed by separating the drug from any additional components.
  • the present invention is further directed towards a novel transdermal delivery device comprising ketamine and abuse deterrent agents.
  • Abuse deterrent agents are employed because they have one or more of the following properties: (1) unpalatable bitterness or other repulsive tastes in the mouth (i.e., bittering agents); (2) formation of gel upon mixing with the extraction solvents (i.e., gel forming agents); (3) severe irritation when injected (i.e.
  • irritants e.g., irritants
  • mood depression e.g., droperidol
  • CNS central nervous system
  • acute gastrointestinal, cardiac or respiratory effects e.g., violent nausea or vomiting
  • repugnant smells if not used as instructed e.g., a violent nausea or vomiting
  • repugnant smells if not used as instructed
  • sleep inducing thereby causing the abuser to miss or be made unaware of the euphoria
  • deactivation or degradation of the active ingredient i.e., strong oxidation agents (such as hydrogen peroxide), strong acid, or strong base, and/or antagonists
  • the abuse deterrent agent is employed at about 0.01-10% by weight of the final formulation, preferably about 0.1-4% by weight, and most preferably about 0.1-0.5% by weight.
  • the abuse deterrent agents can be included in the adhesive drug matrix or in a separate abuse deterrent layer (also referred to as the abuse deterrent matrix).
  • the abuse deterrent layer may be comprised of a combination of polymer and the abuse deterrent agent. Additionally, the abuse deterrent layer can be the abuse deterrent agent itself because many of the recited polymers also act as gel forming agents.
  • Suitable polymers include, but are not limited to, one or more pharmaceutically acceptable polymers that will undergo an increase in viscosity upon contact with a solvent.
  • Preferred polymers include polyethylene oxide, polyvinyl alcohol, hydroxypropyl methylcellulose, carbomers (carbopol), polyvinylpyrrolidone (PVP), and/or other cellulose polymers.
  • the polymer includes polyethylene oxide.
  • the polyethylene oxide can have an average molecular weight ranging from about 300,000-5,000,000, and more preferably from about 600,000-5,000,000, and most preferably at least about 5,000,000.
  • the polyethylene oxide is a high molecular weight polyethylene oxide.
  • suitable, commercially available polyethylene oxide polymers include Poly ox®, WSRN-1105 and/or WSR coagulant, available from Dow Chemical.
  • the preferred weight range of the polymer is from about 1-40% by weight of the final formulation, and the most preferred range of the polymer is from about 2- 10% by weight of the final formulation.
  • bittering agents are pharmaceutically acceptable bitter substances that create a bitter taste or effect when administered nasally (snorted), orally, buccally or sublingually, making consumption difficult.
  • the bittering agents that may be employed in the present invention include, but are not limited to, sucrose octaacetate (used as a denaturant for alcohol) (e.g., SD- 40), denatonium saccharide, denatonium benzoate, caffeine, quinine (or a quinine salt such as quinine sulfate), bitter orange peel oil, and other botanical extract ingredients, such as pepper extract (cubeb), capsicum, and the like.
  • bittering agents are sucrose octaacetate, denatonium benzoate (Bitrex), and denatonium saccharide (four times more bitter than denatonium benzoate) because they are extremely bitter even at low concentrations and are essentially non-toxic.
  • the bittering agent is employed at about 0.01-10% by weight of the final formulation, preferably about 0.1-4% by weight, and most preferably about 0.1-0.5% by weight.
  • Gel forming agents are employed to form a gel structure upon mixing with the extraction solvents and, thus, provide abuse deterrent properties.
  • gel forming agents are compounds that upon contact with a solvent (e.g., water or alcohol), absorb the solvent and swell, thereby forming a viscous or semi-viscous substance that significantly reduces and/or minimizes the amount of free solvent which can contain an amount of solubilized drug, and which minimizes what can be drawn into a syringe for injection (i.e. , IV or intramuscular).
  • the gel can also reduce the overall amount of drug extractable with the solvent by entrapping the drug in a gel matrix.
  • the gel forming agent can be in a segregated abuse deterrent layer laminated to the adhesive drug matrix.
  • the gel forming agents that may be employed include, but are not limited to, ethyl cellulose, cellulose acetate, cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, cellulose ether, cellulose ester, cellulose ester ether, acrylic resins comprising copolymers synthesized from acrylic and methacrylic acid esters, the acrylic polymer may be selected from the group consisting of acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethylmethacrylates, cyanoethyl methacrylate, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, poly(methacrylic acid anhydride), glycidy
  • the gel forming agent includes polyvinyl alcohol with a molecular weight ranging from about 20,000-200,000, specific gravity ranging from about 1.19-1.31, and viscosity ranging from about 4-65 cps.
  • the polyvinyl alcohol used in the formulation is preferably a water-soluble synthetic polymer represented by — (— C2H4 O— )n— , where n can range from about 500-5,000.
  • suitable, commercially available polyvinyl alcohol polymers include PVA, USP, available from Spectrum Chemical Manufacturing Corporation, New Brunswick, N.J. 08901.
  • the gel forming agent includes hydroxypropyl methyl cellulose (Hypromellose) with a molecular weight ranging from about 10,000- 1,500,000, typically from about 5000-10,000 (i.e., low molecular).
  • the specific gravity of the hydroxypropyl methyl cellulose ranges from about 1.19-1.31, with an average specific gravity of about 1.26.
  • Viscosity of the hydroxypropyl methyl cellulose is about 3600-5600 cPs.
  • the hydroxypropyl methylcellulose used in the formulation can be a water-soluble synthetic polymer. Examples of suitable, commercially available hydroxypropyl methylcellulose polymers include Methocel K100 LV and Methocel K4M, available from Dow chemicals.
  • the gel forming agent includes hydrophilic polymers, such as hydrogels, which provides viscosity to the dosage form upon tampering.
  • hydrophilic polymers such as hydrogels
  • a solvent e.g., water or saline
  • the gel forming agent can include carbomers, having a molecular weight ranging from 700,000-4,000,000 and viscosity ranging from about 4000-39,400 cPs.
  • Carbomer is preferably employed in the present invention from about 1-40% by weight of the final formulation, and most preferably from about 2-10% by weight.
  • suitable, commercially available carbomers include carbopol 934P NF, carbopol 974P NF, and carbopol 97 IP NF, available from Lubrizol.
  • Irritants are pharmaceutically inert compounds that induce irritation to the mucous membranes of the body (i.e. , nasal, mouth, eye, intestine, urinary tract).
  • the irritants that may be employed in the present invention include, but are not limited to surfactants, such as sodium lauryl sulfate (SLS), poloxamer, sorbitan monoesters and glyceryl monooleates, as well as spicy ingredients, and others.
  • SLS sodium lauryl sulfate
  • poloxamer poloxamer
  • sorbitan monoesters sorbitan monoesters
  • glyceryl monooleates as well as spicy ingredients, and others.
  • the irritants are preferably employed at about 0.01-10% by weight of the final formulation, preferably 0.01-10% by weight, and most preferably about 0.1- 5% by weight.
  • the irritant can deter abuse upon tampering with the transdermal delivery device. For example, if an abuser extracts and dries the ketamine, then the irritant is exposed and discourages inhalation of the ketamine mixed with the irritant, as inhalation (e.g., via snorting through the nose) will induce pain and/or irritation of the abuser's mucous membrane and/or nasal passageway tissue.
  • the pressure sensitive adhesive (PSA) employed in Examples 1, 2, and 4 was DuroTak 387-2052 (supplied by Henkel Adhesives).
  • the PSA employed in Example 3 was DuroTak 87-2677 (Henkel Adhesives).
  • the PSA employed in Example 5 was Duro-Tak 87-4098 (Henkel Adhesives).
  • Persons skilled in the art will understand that other known pressure sensitive adhesives can be readily employed with the transdermal delivery devices of the present invention.
  • the skin permeation enhancer employed in Examples 4 and 5 was diethylene glycol monoethyl ether, sold under the tradename Transcutol P. Persons skilled in the art will understand that other known skin permeation enhancers can be readily employed with the transdermal delivery devices of the present invention.
  • crystallization inhibitor in Examples 3 and 4 was polyvinyl pyrrolidone-co-vinyl acetate, sold under the tradename Kollidon VA 64 (BASF Corporation).
  • the crystallization inhibitor employed in Example 5 was polymathacrylate-based polymer, sold under the tradename Plastoid B (Evonik Corporation). Persons skilled in the art will understand that other known crystallization inhibitor enhancers can be readily employed with the transdermal delivery devices of the present invention.
  • Figure 5 is a comparison of the plasma concentration-time profile of a 0.5 mg/kg dose of ketamine in a human subject following: (1) a 40-minute single IV administration; and (2) administration of a 24 hour transdermal delivery device of the present invention according Example 2 (a 9.4 cm 2 transdermal delivery device with a 3.75 mg/cm 2 permeation rate).
  • Convolution analysis was applied in accordance with the pharmacokinetic parameters set forth in Fanta, et al., Eur. J. Clin. Pharmacol, 71 :441-447 (2015).
  • the transdermal delivery device according to Example 2 exhibits a lower Cmax, preferably less than about 30%, and more preferably less than about 20%, of the Cmax from an equivalent IV dosage.
  • Figure 6 discloses ketamine plasma concentration-time profiles for three sizes (10, 100 and 300 cm 2 ) of the once-a-day transdermal delivery device of the present invention according to Example 2.
  • Figure 7 discloses ketamine plasma concentration-time profiles for three sizes (10, 100 and 300 cm 2 ) of the three times-a-day transdermal delivery device of the present invention according to Example 2.
  • Skin permeation enhancers are incorporated in the transdermal delivery devices of the present invention to ensure that sufficient ketamine can penetrate through skin.
  • Skin permeation studies were performed on the transdermal delivery devices prepared according to Examples 1-5 using Franz diffusion cells maintained at 37° C.
  • the receptor medium was phosphate buffered saline at pH 7.4, the receptor volume was 12 ml and the permeation area was 1.767 cm 2 .
  • Human cadaver skin was used and the tests were performed in triplicate.
  • a lxl inch transdermal delivery device was placed onto the donor side of the skin diffusion cells, adhered onto the skin, and the experiment was initiated with the receptor medium being continuously mixed (stirring at 600 rpm).
  • Figure 8 depicts in vitro skin permeability of the transdermal delivery devices of the present invention according to Example 2 as shown by the Franz Diffusion Cell model.
  • Drug crystallization will retard drug release and skin permeability, reducing the efficacy of the transdermal delivery device.
  • Drug crystals should not be formed in the transdermal delivery device over a period approximating the shelf life, i.e., for about 6 months or greater.
  • Examples 1 and 2 showed instability, i.e. drug crystals were formed in the adhesive drug matrix 4-7 days after preparation of the transdermal delivery devices.
  • Examples 3, 4 and 5, were found to be stable for at least 4 weeks, i.e. within this period, no crystals were formed.
  • Example 5 was found to be stable for at least 6 months at conditions of 25° C, 60% RH.
  • Table 8 reports the stability data for the transdermal delivery devices prepared according to Examples 1-5.
  • Figure 9 depicts the stability of ketamine in a transdermal delivery device according to Example 5 after 0 months and after 6 months.
  • Example 5 was modified to prepare a transdermal delivery device that contains 5 mg of denatonium benzoate and 200 mg of ketamine in the adhesive drug matrix.
  • In vitro skin permeation studies showed that no denatonium benzoate permeated the skin (because of its large molecular weight (447 DA) and the high melting point (170° C). Nevertheless, ketamine showed excellent skin permeability (0.8 mg/cm 2 in 24 hours), indicating that incorporation of an abuse deterrent agent, such as denatonium benzoate did not affect the skin permeation of ketamine.
  • transdermal delivery devices prepared according to Example 6 were soaked for 60 minutes in 100 ml of three different media: (1) 40% ethanol; (2) 70% isopropyl alcohol; and (3) acetone. All three media were assayed for ketamine and denatonium benzoate using HPLC. More than 50% of the original ketamine and more than 50% of the denatonium benzoate were found in the media (i.e. , the bittering agent extracted proportionally to the amount of ketamine extracted), indicating the effectiveness of using denatonium benzoate as an abuse deterrent agent in the present invention.
  • Example 7 comprises a transdermal delivery device with an abuse deterrent agent in a segregated layer.
  • the abuse deterrent agent employed in Example 7 is a gelling agent which reacts with common solvents (e.g., water and alcohol) used to extract and abuse the ketamine in the transdermal delivery device.
  • the thickness of the adhesive-drug layer and the abuse deterrent layer are both about 2 to about 5 mils.
  • the transdermal delivery device according to Example 7 is prepared in a two-step process.
  • An abuse deterrent agent such as Bitrex and sodium lauryl sulfate (SLS) may be incorporated into the abuse deterrent layer, preferably from about 0.01-5% by weight of the final formulation, and most preferably from about 0.05-0.5% by weight of the final formulation.
  • SLS sodium lauryl sulfate
  • the adhesive drug matrix layer is prepared by casting the adhesive drug matrix mix directly on the abuse deterrent layer (prepared in step 1), or onto a release liner and then laminated to the abuse deterrent layer.
  • DuroTak 87-4098 is weighed into a 100 ml beaker, and then mixed at low speed. Next, the Kollidon VA 64 and the ketamine are added to the mixer. The batch is mixed until all ingredients are dissolved. Then wet films are prepared at 3 mils thickness using a film casting applicator on release liner, such as 3M's 9744. The wet coating is air dried for 1 hour, and then oven dried at 60° C for 10 min. Finally, the laminate is dried onto the abuse deterrent layer, which was subsequently coated on 3M's backing Scotpak 1012.
  • the laminated sheet can be die-cut into transdermal delivery devices of various sizes, such as 10 cm 2 , 20 cm 2 , 100 cm 2 , 300 cm 2 to obtain the desired dosages of the drug.
  • a transdermal delivery device is prepared with apomorphine, an emetic, in the abuse deterrent layer. After solvent extraction by an abuser, the emetic can cause severe nausea if injected, snorted, or inhaled.
  • Apomorphine is preferably employed in the present invention from about 0.05-5% by weight of the final formulation, and most preferred from about 0.1-2% by weight.
  • Example 9 was prepared in accordance with Example 7, with the exception that the abuse deterrent agent is capsaicin. After being dissolved in solvents by an abuser, the ketamine solution containing capsaicin will cause a torturous buming sensation if snorted or inhaled, thereby reducing the abuse potential of the transdermal drug delivery device.
  • the abuse deterrent agent is capsaicin.
  • Table 11 provides additional techniques by which the abuse deterrent agents can be employed in the transdermal delivery device of the present invention.
  • the adhesive-drug layer comprises an adhesive and 200 mg of ketamine
  • the abuse deterrent layer comprises a gel forming agent comprising 7 mg of SLS and 5 mg of denatonium benzoate.

Abstract

TThe present invention is directed to a transdermal delivery device comprising ketamine and formulations thereof. The present invention is also directed to a transdermal delivery device comprising ketamine for the treatment of major depressive disorder (MDD) and/or pain. The present invention is further directed to a transdermal delivery device comprising ketamine and abuse deterrent agents.

Description

KET AMINE TRANSDERMAL DELIVERY SYSTEM
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. provisional patent application No. 62/185,573 filed June 27, 2015.
FIELD OF THE INVENTION
The present invention is directed to transdermal delivery devices comprising ketamine and formulations thereof. The present invention is also directed to transdermal delivery devices comprising ketamine for the treatment of major depressive disorder (MDD) and/or pain. The present invention is further directed to transdermal delivery devices comprising ketamine and abuse deterrent agents.
BACKGROUND OF INVENTION
Major depressive disorder (MDD) is a disabling psychiatric illness. Lifetime prevalence of MDD is approximately 16%. Kessler et al, JAMA, 289(23):3095-105 (2003). There are three primary classes of antidepressants that are commonly prescribed for MDD: (1) monoamine oxidase inhibitors (MAOIs); (2) tricyclics; and (3) serotonin-norepinephrine reuptake inhibitors (SNRIs) and selective serotonin reuptake inhibitors (SSRIs). There are significant limitations with the use of current antidepressants, including limited efficacy, delayed onset of action, and adverse side effects. Additionally, antidepressants have been found to be only about 20-30% more effective than placebo. The delay of onset varies from weeks to months, which may result in adverse events, including but not limited to increased vulnerability to suicide, decrease in compliance, and increase in social and economic burden. Common side effects of these antidepressants include nausea, insomnia, anxiety, weight- loss/gain, drowsiness, headache, loss of sex drive, and/or blurred vision. Perm and Tracey, Ther Adv. Psychopharmacol., 2(5): 179-188 (2012).
Pain can present as a disabling physical illness. One type of pain, neuropathic pain, is a complex chronic pain state often accompanied by tissue injury. The occurrence of pain with neuropathic characteristics is about 6.9-10% of the general population. Hecke et al , Pain, 155(4):654-62 (2014). Symptoms of neuropathic pain include spontaneous burning, shooting pain, hyperalgesia, and allodynia. Patients with neuropathic pain often have conditions that are associated with other significant health issues, including depression, sleep problems, and loss of independence. Bouhassira ei a/., Pain., 136(3):380-7 (2008). Neuropathic pain can be caused by a variety of mechanisms, including infection, central or peripheral nerve injury, stroke, multiple sclerosis, diabetes mellitus, sarcoidosis, toxic agents (e.g., alcohol or chemotherapy), inherited or genetic neuropathy, and Complex Regional Pain Syndrome (CRPS). CRPS is an intractable form of pain, often resistant to a variety of conventional therapies. Correll et al, Pain Med., 5(3):263-75 (2004). Neuropathic pain is difficult to treat, with only about 40-60% of patients achieving partial relief. Treatment for neuropathic pain includes antidepressants, anticonvulsants, and/or topical pain management medications. Niesters et al , Expert Opin. Drug Metab. Toxicol, 8(11): 1409-17 (2012); Dworkin ei al, Pain, 132(3):237-51 (2007).
Ketamine is a non-competitive, N-methyl-D-aspartate (NMDA) receptor antagonist, indicated for treatment as an anesthetic, sedative, and analgesic. Ketamine has been demonstrated to be an effective antidepressant, with rapid onset (within about 2 hours of administration) and sustained antidepressant effect (from days to in some cases a week or two after administration). Berman et al, Biol. Psychiatry, 47(4):351-54 (2000). The NMDA receptor pathway plays an important role in pain, including neuropathic pain. Animal studies and human clinical studies have shown the efficacy of ketamine in the treatment of chronic neuropathic pain. Correll et al , Pain Med. 5(3):263-75 (2004); Sigtermans et al, Pain, 145(3):304-11 (2009).
Ketamine is a racemic mixture containing R-ketamine and S-ketamine. It is generally believed that the anesthetic and/or antidepressant effect of ketamine is mainly through the action of S-ketamine because in vitro S-ketamine has about a 4-fold greater affinity than the R-ketamine on NMDA receptor binding. However, animal model studies have suggested that R-ketamine is more effective as an antidepressant than S-ketamine. In addition, R-ketamine was shown to be free of psychotomimetic side effects and abuse liability. Yang et al , Transl. Psychiatry, 5(e632): l-l l (2015). The present invention is directed toward administration of the racemic mixture of ketamine; however, embodiments containing the R-ketamine or the S- ketamine enantiomers are within the scope of the present invention.
Ketamine is also a known dissociative anesthetic that has gained popularity as a drug of abuse, and may be referred to illicitly as "K" or "Special K". Ketamine is reported to distort perceptions of sight and sound, and make the user feel disconnected. The 2011 "Monitoring the Future" (MTF) study reports the annual use of ketamine among 8th, 10th, and 12th graders as being 0.8%, 1.2%, and 1.7%, respectively. Johnston, et al, 2012, Monitoring the future national results on adolescent drug use: Overview of key findings, 2011, Ann Arbor: Institute for Social Research, The University of Michigan. Illicit ketamine can be distributed as a dried powder or as a liquid, mixed with beverages, and/or added to smokable materials (such as marij uana or tobacco). As a powder, ketamine can be snorted or pressed into tablets, sometimes in combination with other drugs, including 3,4-methylenedioxymethamphetamine (MDMA, referred to illicitly as "ecstasy"), amphetamine, methamphetamine, cocaine, and/or carisoprodol. On August 12, 1999 ketamine became a Schedule III non-narcotic substance under the Controlled Substances Act. Consequently, there is a need to develop abuse deterrent mechanisms to reduce the risk of ketamine abuse.
IV administration of ketamine presents numerous challenges. First, the patient incurs increased costs to receive IV administration. Second, IV administration is inconvenient for the patient, and may lead to reduced compliance. Third, the rapid initial rise in ketamine plasma concentrations following IV administration to the maximum plasma concentration (Cmax) can cause adverse side effects, including drug toxicity, psychotomimetic problems, and increased potential for addiction. Moreover, because ketamine has a short half-life (about 2 hours), this immediate release delivery of ketamine by IV administration may result in little to no ketamine remaining in plasma after about 4-8 hours, necessitating frequent and repeated dosing to maintain therapeutic plasma levels. Fourth, without additional safeguards, IV administration of ketamine may be susceptible to abuse.
An intranasal formulation of the S-enantiomer of ketamine, esketamine, is under development and in clinical study by Janssen. US 2013/0236573 Al, Singh et al, Esketamine For The Treatment of Treatment-Refractory Or Treatment-Resistant Depression. However, intranasal delivery of ketamine presents numerous challenges. It suffers from many of the same immediate release issues faced by IV administration of ketamine, namely, rapid onset of maximum concentration (Tmax), high Cmax, increased risk of side effects like drug toxicity, and the need for frequent and multiple dosing to maintain therapeutic plasma concentrations. Frequent administration of intranasal ketamine may increase the risk of irritating and damaging the nasal epithelium, which in turn may reduce patient compliance. Also, intranasal administration is associated with high variability in absorption among subjects. Kublik et al, Adv. Drug Deliv. Rev. 29: 157-77 (1998). Further, the rapid rise in ketamine plasma concentration following intranasal administration may cause adverse side effects, such as drug toxicity. Moreover, intranasal delivery of ketamine, without additional safeguards, is highly susceptible to abuse. Other routes of administration of ketamine, including parenteral administration of ketamine (e.g., subcutaneous, intramuscular, etc.) suffer from many of these same challenges.
While oral administration (i.e. , tablet or capsule) is typically convenient for the patient, the metabolic and pharmacokinetic properties of ketamine make oral administration less suitable. Ketamine has a high systemic (primarily hepatic) clearance of about 19 ml/min»kg, a rate which approaches liver plasma flow. Thus, ketamine is subject to substantial pre- systemic metabolism, or first-pass effect, in the liver and gut wall by metabolic enzymes, such as cytochrome P450 enzymes (CYP450). Consequently, the absolute oral bioavailability of ketamine in humans is only about 10-20%. Due to this first-pass effect, there is an increased risk for drug-drug interactions (DDI) with drugs that can inhibit or induce CYP450s. Clements et al. , J Pharm Sci, 71(5):539-42 (1981); Fanta, et al, Eur. J. Clin. Pharmacol, 71 :441-47 (2015); Peltoniemi et al , Basic & Clinical Pharmacology & Toxicology, 111 :325-332 (2012). Moreover, ketamine tablets or capsules are easily abused.
SUMMARY OF THE INVENTION
The present invention is directed to transdermal delivery devices comprising ketamine and formulations thereof. The present invention is also directed to transdermal delivery devices comprising ketamine for the treatment of major depressive disorder (MDD) and/or pain. There are long-felt and unmet medical needs for the treatment of MDD and for the treatment of pain, which are fulfilled by the present invention. The controlled, prolonged, and steady ketamine exposure to humans from the transdermal delivery device of the present invention can reduce adverse side effects compared with other routes of ketamine delivery, including but not limited to intravenous (IV) administration and intranasal spray. Because ketamine has high abuse potential, the present invention is further directed to transdermal delivery devices comprising ketamine and abuse deterrent agents.
The present invention has numerous advantages. Formulations of the transdermal delivery device provide excellent ketamine permeability and stability. The inventors discovered, through in vitro experiments, that ketamine has excellent transdermal permeability properties, which are very important for efficacious clinical use. Additional in vitro experiments demonstrated that adding, for example, crystallization inhibitors to formulations of the present invention resulted in very stable transdermal delivery devices, which is important when making a pharmaceutical product.
Additionally, the present invention provides improved drug metabolism and pharmacokinetic properties compared with other methods of administering ketamine, such as IV and intranasal administration. First, transdermal delivery avoids the aforementioned first- pass effect. Second, it reduces the aforementioned DDI risk. Third, it delivers a sustained in vitro release profile, and therefore, a steadier in vivo plasma concentration versus time profile over a longer period of time. In other words, there is no need for frequent, multiple dosing for days or weeks to maintain therapeutic plasma concentrations of ketamine, as may be the case with, for example, IV infusion. Correll et al, Pain Med. 5(3):263-75 (Sept. 2004). Instead, the present invention can meet the desired prolonged drug absorption profile. For example, administration of a single transdermal delivery device of the present invention can deliver a relatively constant ketamine plasma concentration for up to about 7 days. Fourth, the present invention delivers lower Cmax values for ketamine in the plasma, as well as minimal fluctuation between Cmax and Cmin, thereby reducing adverse side effects, including but not limited to toxicity, psychotropic effects, increased potential for addiction, and lack of therapeutic effect.
The transdermal delivery device of the present invention provides flexibility in dose, dosage release rate, patch size, and duration of application to allow for optimization. Transdermal delivery devices according to the present invention include, but are not limited to, transdermals and dermal patches, topical skin applications such as spray, creams, gels, lotions, dressings and liquid solutions, and other transdermal delivery systems and dosage forms known to persons skilled in the art. For example, these flexible parameters can be adjusted by the formulator and/or the clinician to provide the optimal ketamine plasma concentration-time profile for the individual patient that maximizes efficacy and minimizes adverse side effects. Therefore, the ketamine transdermal delivery device of the present invention is particularly effective for the treatment of MDD and pain.
Moreover, the present invention improves convenience and compliance compared with other forms of administering ketamine. For example, administration of the transdermal delivery device once or twice a week is more convenient than, for example, multiple daily doses of immediate release forms of ketamine. Immediate release ketamine or immediate release ketamine formulation means administration of ketamine that is not extended, controlled, delayed or prolonged. Dose equivalent with means that the total dosage of drug administered between the compared items is the same. Also, the present invention is less invasive and less costly than IV administration. The present invention causes less irritation and is less invasive than intranasal administration. Further, the present invention is less likely to cause drug toxicity than immediate release forms of drug delivery.
The present invention also has advantages with respect to abuse deterrence. The transdermal delivery device itself may serve as an abuse deterrent because it is more difficult to abuse directly, such as by biting or swallowing the device. Specifically, the ketamine is incorporated into a polymeric matrix together with other excipients, including, but not limited to skin permeation enhancers, humectants, plasticizers, buffers, antioxidants, and combinations thereof, each of which may inhibit ketamine extraction for abuse. Nevertheless, specific, additional abuse deterrent agents can be added to formulations of the present invention to further deter abuse.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a ketamine transdermal delivery device without abuse deterrent properties.
Figure 2 is a transdermal delivery device comprising ketamine with abuse deterrent properties.
Figure 3 is an alternate embodiment of a transdermal delivery device comprising ketamine and abuse deterrent agents.
Figure 4 is an alternate embodiment of a transdermal delivery device comprising ketamine and abuse deterrent agents.
Figures 5-7 are ketamine plasma concentration versus time profiles in humans for Example 2 of the present invention using different sizes of the transdermal delivery device. These pharmacokinetic profiles are predicted by known convolution methodology using in vitro transdermal permeation data and in vivo intravenous plasma concentration data.
Figure 8 is a graph of the in vitro skin permeation of ketamine in a Franz Diffusion Cell model for the transdermal delivery device according to Example 2.
Figure 9 is a 6-month stability graph of the present invention prepared in accordance with Example 5.
Figures 10-13 are ketamine plasma concentration versus time profiles in humans corresponding to the transdermal delivery devices of Table 2. These transdermal delivery devices are for the treatment of MDD. These pharmacokinetic profiles are predicted by known convolution methodology using in vitro transdermal permeation data and in vivo intravenous plasma concentration data.
Figures 14-17 are graphs of the pharmacokinetic plasma concentrations versus time profiles, corresponding to the transdermal delivery devices of Table 3. These transdermal delivery devices are for the treatment of pain. These pharmacokinetic profiles are predicted by known convolution methodology using in vitro transdermal permeation data and in vivo intravenous plasma concentration data.
DETAILED DESCRIPTION OF THE INVENTION
The active ingredient in the transdermal delivery device of the present invention is preferably employed at about 1-35% by weight of the final formulation (also referred to as 1- 35 weight percent) and most preferably 10-25% by weight of the final formulation. The most preferred active ingredient is ketamine. Additional active ingredients that can be employed in the present invention can be chosen from drugs that counteract the potential adverse effects of ketamine, reduce ketamine addiction potential, and/or enhance ketamine's antidepressant effect and/or pain management effects. All weight percentages provided in the present disclosure are based on the weight of the final formulation, which includes the adhesive-drug layer (or adhesive-drug matrix) and the abuse deterrent layer (or abuse deterrent matrix, which is optional), but not the release liner or the backing film.
The transdermal permeation rate (mg/day) combined with the size of the transdermal delivery device and the duration of application of the transdermal delivery device determines the plasma concentration of the drug. The transdermal permeation rate of the ketamine transdermal delivery device of the present invention will preferably be about 0.1-30 mg/day/cm2 of the transdermal delivery device, and most preferably about 0.5-5 mg/day/cm2. The size of transdermal delivery device will preferably be about 5 -300 cm2. The duration of application of the transdermal devices will preferably be about 8-168 hours. Combinations of these preferred ranges provide prolonged plasma concentrations of ketamine ranging from about 0.4-3850 ng/ml. The plasma concentrations of ketamine will reach near steady-state at about 8 hours after administration and will be sustained for the duration of the application. Preferred transdermal permeation rates, transdermal delivery device sizes, and durations of applications for the transdermal delivery devices of the present invention are set forth in Table 1.
TABLE 1
Figure imgf000009_0001
The plasma concentrations of ketamine versus time are calculated based on the reported pharmacokinetic parameters of ketamine in humans. Ketamine follows a three-compartment model with the parameters for a 70-kg human as follows: clearance = 79.8 (liter/hour); VI = 133 liter; and micro constants ki2 = 0.174 hour"1, ki3 = 1.18 hour"1, k2i = 0.124 hour"1, k3i = 1.59 hour"1. Fanta, et al, Eur. J. Clin. Pharmacol, 71 :441-447 (2015). There is variability among human populations and individuals, such that the pharmacokinetics of each human are not the same, and for certain populations and for some individuals, the pharmacokinetics can deviate significantly. Plasma concentrations provided by any given dosage strength for the transdermal delivery device of the present invention can vary from individual to individual.
For antidepressant effect (i.e. , the treatment of MDD) with minimized adverse side effects, the preferred plasma concentration range of ketamine is from about 10-200 ng/ml, and the most preferred plasma concentration range is from about 10-100 ng/ml.
Table 2 provides ranges for the most preferred transdermal permeation rates, transdermal delivery device sizes, and duration of application of the transdermal delivery devices for antidepressant effect (i.e. , the treatment of MDD) with minimized adverse side effects. These ranges are chosen to provide prolonged plasma concentrations of ketamine ranging from about 10 -200 ng/ml for about 8-168 hours, and the most preferred plasma concentrations are from about 10-100 ng/ml. Variations on the transdermal delivery devices comprising ketamine designed for the treatment of MDD will contain from about 8.3-200 mg and be applied for about 8 hours, from about 25-600 mg and be applied for about 24 hours, from about 87.5-2100 mg and be applied for about 84 hours, and from about 175-4200 mg and be applied for about 168 hours.
Transdermal delivery devices for the treatment of MDD are prepared with dosage strengths from about 8.3-200 mg, and are designed to be applied for about 8 hours, which will provide a plasma concentration of ketamine from about 11-257 ng/ml. An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects. The reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 8.3-100 mg of ketamine, designed to be applied for 8 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
Transdermal delivery devices for the treatment of MDD according to the present invention are prepared with dosage strengths from about 25-600 mg and are designed to be applied for about 24 hours, which will provide a plasma concentration of ketamine from about 11 -257 ng/ml. An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects. The reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 25-300 mg of ketamine, designed to be applied for 24 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
Transdermal delivery devices for the treatment of MDD according to the present invention are prepared with dosage strengths from about 87.5-2100 mg, and are designed to be applied for about 84 hours, which will provide a plasma concentration of ketamine from about 11 -257 ng/ml. An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects. The reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 87.5-1050 mg, designed to be applied for about 84 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
Transdermal delivery devices for the treatment of MDD according to the present invention are prepared with dosage strengths from about 175-4200 mg, and are designed to be applied for about 168 hours, which will provide a plasma concentration of ketamine from about 11 -257 ng/ml. An alternate embodiment of the present invention for the treatment of MDD is designed to provide reduced adverse side effects. The reduced adverse side effects are provided by a transdermal delivery device according to the present invention prepared with dosage strengths of ketamine from about 175-2100 mg, designed to be applied for about 168 hours, and to provide a plasma concentration of ketamine from about 11-128 ng/ml.
TABLE 2
Figure imgf000011_0001
For pain management, with minimized adverse events, the preferred plasma concentrations ranges of ketamine are from about 50-1000 ng/ml, and the most preferred plasma concentration is about 500 ng/ml.
Table 3 provides ranges for transdermal permeation rates, transdermal delivery device sizes, and duration of application of the transdermal delivery devices for pain management. These ranges are chosen to provide prolonged plasma concentrations of ketamine ranging from about 50-1000 ng/ml, and for about 8-168 hour, and the most preferred plasma concentration is about 500 ng/ml. Transdermal delivery devices comprising ketamine designed for pain management will contain about 40-500 mg and be applied for about 8 hours, from about 120- 1500 mg and be applied for about 24 hours, from about 420-5250 mg and be applied for about 84 hours, and from about 840-10500 mg and be applied for about 168 hours.
Transdermal delivery devices for the treatment of pain according to the present invention are prepared with dosage strengths from about 40-500 mg, are designed to be applied for about 8 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml. Transdermal delivery devices for the treatment of pain according to the present invention are prepared with dosage strengths from about 120-1500 mg, are designed to be applied for about 24 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
Transdermal delivery devices for the treatment of pain according to the present invention are prepared with dosage strengths from about 420-5250 mg, are designed to be applied for about 84 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
Transdermal delivery devices for the treatment of pain according to the present invention are prepared with dosage strengths from about 840-10,500 mg, are designed to be applied for about 168 hours, and will provide a plasma concentration of ketamine from about 51-642 ng/ml.
TABLE 3
Figure imgf000012_0001
Preferably, the transdermal delivery device of the present invention will be administered once a day, twice a week, or once a week. The dosing regimen of the present invention is not limited to the examples provided in Tables 2 and 3 for antidepressant effect and pain management. In accordance with the need of the patient and as determined by the physician, the dose frequencies, device size, and/or dosage strength can be adjusted. For example, the application of the transdermal delivery device can be for a duration shorter than 8 hours, such as 4 hours. The ketamine plasma concentrations at about 4 hours after administration will be about 80% of the plasma concentration at about 8 hours, which can provide effective anti-depressant and/or pain management, depending on the need of the patient. The plasma drug concentrations profiles in Table 4 and illustrated in Figs. 6-7, are exemplary of the plasma profiles of the present invention. The plasma drug concentration profiles in Tables 2 and 3 and illustrated at least in 10-17, are further exemplary of the plasma profiles of the present invention. These plasma profiles rise slowly and are maintained at a relatively constant level for a prolonged period of time. On the contrary, IV and intranasal ketamine will generally provide a Cmax approximately 3 to 10 times higher than the Cmax provided by a transdermal delivery device of the present invention (at equivalent doses), while the area under the curve (AUC) is constant (e.g. , Figure 5). Additionally, the prolonged and steady administration of ketamine provided by the transdermal delivery device of the present invention exhibits minimal fluctuations in plasma concentration relative to multiple doses of IV or intranasal administration of ketamine. This reduction or minimization of plasma fluctuations in turn reduces the occurrence of adverse side effects resulting from under and over medication. Consequently, the plasma profile provided by the present invention is improved and may result in better therapeutic outcomes and greater patient compliance.
The structure and packaging of the transdermal delivery device of the present invention are prepared in accordance with methods and techniques known to persons skilled in the art. The primary components are the backing layer, the adhesive-drug layer (or adhesive-drug matrix), the abuse deterrent layer (or abuse deterrent matrix) (optional), and the release liner.
The backing layer may be comprised of polymeric films such as polyester (PET) or polyethylene (PE) films that support the adhesive drug matrix and protect the transdermal delivery device from the environment. The preferred thickness range for the backing film is from about 2-5 mils (1 mil equals 1/1000 of an inch), and the most preferred thickness range of the backing layer is from about 3-4 mils thick.
The adhesive in the adhesive-drug layer may be a pressure sensitive adhesive (PSA). Tan et al , Pharm Sci. & Tech Today , 2:60-69 (1999). Useful PSAs in transdermal delivery systems include, but are not limited to, polyisobutylenes (PIB), silicone polymers, and acrylate copolymers, such as acrylic pressure sensitive adhesives, including Duro-Tak 87-2516, 87- 2852 and 87-2194, manufactured by Henkel Adhesives. PIBs are elastomeric polymers that are commonly used in PSAs, both as primary-base polymers and as tackifiers. PIBs are homopolymers of isobutylene and feature a regular structure of a carbon-hydrogen backbone with only terminal unsaturation. PIBs are marketed under the trade name Oppanol by BASF. The silicone polymers are a high molecular weight polydimethylsiloxane that contains residual silanol functionality (SiOH) on the ends of the polymer chains. Silicone PSAs for use in pharmaceutical applications are available from Dow Corning Corporation, for example under the trade name of BIO-PSA. The PSA is preferably employed at about 30-90% by weight of the final formulation, and most preferably about 40-60% by weight of the final formulation.
The release liner can be manufactured in the desired size for the present invention. The release liner may be comprised of silicone or fluoro-polymer coated polyester film. The release liner protects the transdermal delivery device during storage and is removed before its use. Silicone-coated release liners are manufactured by Mylan Corporation, Loparex Corporation, and 3M's Drug Delivery Systems. The fluoro-polymer coated release liners are manufactured and supplied by 3M's Drug Delivery Systems and Loparex. The preferred thickness of the release liner is about 2-10 mils, and most preferably about 3-5 mils.
Additional drugs can be incorporated in the transdermal delivery device to counteract adverse effects, and/or to enhance the antidepressant or pain management effect of ketamine. Examples for enhancing antidepressant effect include, but are not limited to, antagonists of group II metabotropic glutamate receptors, such as LY341495, Podkowa et al., Psychopharmacology (Berl) (Jun 11 2016). Examples for reducing side effects with ketamine, especially psychotomimetic and sympathomimetic, include, but not limited to, coadministration of alpha-2 agonists such as clonidine. Lenze, World J Biol Psychiatry, 17(3):230-8 (2016). If an additional drug is employed in the present invention, it is preferably employed at about 0.1-20% by weight of the final formulation, and most preferably about 1- 5% by weight of the final formulation.
Additional components can be added to the transdermal delivery device of the present invention to optimize it. Skin permeation enhancers are employed to enhance the skin permeability of the drug through the skin. Skin permeation enhancers that may be employed in the present invention include, but are not limited to, sulphoxides (e.g. dimethylsulphoxide, DMSO), Azones (e.g. laurocapram), pyrrolidones (e.g. 2-pyrrolidone, 2P), alcohols and alkanols (ethanol, or decanol), glycols (e.g. propylene glycol (PG)), surfactants and terpenes. Williams et al. , Adv Drug Deliv Rev. 27;56(5):603-l 8 (2004). The skin permeation enhancers are preferably employed at about 1-20% by weight of the final formulation, and most preferably about 4-10% by weight of the final formulation.
Humectants are employed to keep the transdermal delivery device hydrated and/or to reduce the loss of moisture. The humectants that may be employed in the present invention include, but are not limited to, propylene glycol, glycerol, urea, polyvinylpyrrolidone (PVP), vinylpyrrolidone-vinyl acetate copolymers, and copolymers of PVP (e.g., BASF's Kollidon K30, K12, Kollidon VA 64, or Kollidon CL-M, magnesium silicate, and silica. The humectants are preferably employed at about 2-20% by weight of the final formulation and most preferably about 5-10% by weight of the final formulation.
Plasticizers are employed in transdermal drug delivery systems to obtain desirable mechanical properties, such as to improve the film forming properties and the appearance of the film, to decrease the glass transition temperature of the polymer, to prevent film cracking, and to increase film flexibility. The plasticizers that may be employed in the present invention include, but are not limited to, phthalate esters, phosphate esters, fatty acid esters, and glycol derivatives. The plasticizers are preferably employed at about 2-20% by weight of the final formulation and most preferably about 5-10% by weight of the final formulation. Designing and Characterization of Drug Free Transdermal delivery devices for Transdermal Application, International Journal of Pharmaceutical Sciences and Drug Research, Vol. 2, No. 1 , pp. 35-39 Bharkatiya, M ; Nema, R.K. & Bhatnagar, M. (2010) Wypch, G. (2004) and Handbook of Plasticizers, Chem Tec, 437-440, ISBN 1-895198-29-1, Ontario, Canada.
Antioxidants are employed to prevent drug degradation by oxidation. Antioxidants that may be employed in the present invention include, but are not limited to, butylated hydroxyanisole (BHA), butylhydroxy toluene (BHT), tert-Butylhydroquinone, ascorbic acid, and tocopherols. The antioxidants are preferably employed at about 0.01 -5% by weight of the final formulation and most preferably about 0.1-1.0% by weight of the final formulation.
Anti-irritants are employed to provide alleviation or prevention of skin irritation, and to assist in the release of the active ingredients. Anti-irritants that may be employed in the present invention include, but are not limited to, aloe, arnica, chamomile, cucumber, menthol, mugwort, oat, zinc oxide, drug release modifiers such as chitosan, cellulose-based polymers, silicon dioxides, and polymethacrylates.
Other suitable excipients useful in the preparation of transdermal delivery devices are within the knowledge of those skilled in the art, and can be found in the Handbook of Pharmaceutical Excipients, (7th ed. 2012), the entire content of which is hereby incorporated by reference. Figure 1 is an embodiment of the transdermal delivery device in which the backing film
(1) is affixed atop the adhesive drug matrix (2), which is supported by a release liner (3). The adhesive drug matrix contains the drug and adhesive, as well as enhancers, humectants, plasticizers, antioxidants, pH modifiers, crystallization inhibitors, and other ingredients that aid in drug release and permeation through skin, and in maintaining drug stability.
Figure 2 is an example of a transdermal delivery device that contains an abuse deterrent agent (4) that is not skin permeable, drug (5) dissolved in the adhesive-drug matrix (2), a transdermal delivery device backing film (1), and a release liner (3).
Figure 3 is a transdermal delivery device that contains a backing film (1), an abuse deterrent layer (6), an adhesive-drug matrix (2), and a release liner (3). The abuse deterrent layer is capable of releasing an abuse deterrent agent upon tampering with the transdermal delivery device. In an embodiment in which the abuse deterrent layer comprises a gel forming agent, the gel forming agent can form a gel solution upon extraction.
Figure 4 is an embodiment of a prolonged use, e.g., 7-day transdermal delivery device, with abuse deterrent agents in a segregated, abuse deterrent layer. Figure 4 shows the backing film (1), and overlay adhesive layer (7), an abuse deterrent layer (6), the adhesive-drug layer
(2) , and a release liner (3). The overlay adhesive layer extends over the outer edges of the drug-adhesive layer (2) and the abuse deterrent layer (6) to provide added adhesion to the skin for prolonged use. Embodiments of the present invention can be prepared with an overlay adhesive layer (7) with or without an intervening abuse deterrent layer (6).
Table 4 provides estimated plasma concentrations for transdermal delivery devices prepared according to the present invention. The plasma concentration are exemplified in Figs. 5, 6 and 7, which are described below in detail.
TABLE 4
Figure imgf000016_0001
23 13 14 138 414 14 138 414
17 14 15 146 439 15 146 439
14 14 15 153 458 15 153 458
11 15 16 158 473 16 158 473
8.9 15 16 162 485 10 102 305
7.4 16 17 165 495 7.5 75 223
6.4 16 17 168 504 5.7 57 169
5.5 16 17 170 511 4.4 44 132
4.9 16 17 173 518 3.5 35 106
4.3 16 17 175 524 2.9 29 87
3.9 17 18 176 529 2.4 24 73
3.5 17 18 178 534 2.1 21 63
3.2 17 18 179 538 1.8 18 54
2.9 17 18 181 542 1.6 16 48
2.6 17 18 182 546 1.4 14 43
2.4 17 18 183 549 1.3 13 38
2.2 17 18 184 552 1.2 12 35
2.0 17 18 185 554 1.0 10 31
1.8 17 19 186 557 0.95 9.5 28
1.7 18 19 186 559 0.86 8.6 26
1.5 12 13 125 375 0.79 7.9 24
1.4 9.0 9.6 96 287 0.72 7.2 21
1.3 7.1 7.6 76 227 0.65 6.5 20
1.2 5.8 6.1 61 184 0.60 6.0 18
1.1 4.8 5.1 51 154 0.55 5.5 16
0.97 4.1 4.3 43 130 0.50 5.0 15
0.88 3.5 3.8 38 113 0.46 4.6 14
0.81 3.1 3.3 33 99 0.42 4.2 13
0.74 2.7 2.9 29 88 0.38 3.8 11
0.68 2.5 2.6 26 78 0.35 3.5 10
0.62 2.2 2.3 23 70 0.32 3.2 9.6
0.57 2.0 2.1 21 64 0.29 2.9 8.8
0.52 1.8 1.9 19 58 0.27 2.7 8.0
0.47 1.6 1.7 17 52 0.24 2.4 7.3
0.43 1.5 1.6 16 48 0.22 2.2 6.7
0.40 1.4 1.5 15 44 0.21 2.1 6.1
0.36 1.2 1.3 13 40 0.19 1.9 5.6
0.33 1.1 1.2 12 36 0.17 1.7 5.1
0.30 1.0 1.1 11 33 0.16 1.6 4.7
0.28 0.95 1.0 10 30 0.14 1.4 4.3
0.25 0.87 0.92 9.2 28 0.13 1.3 3.9
0.23 0.79 0.85 8.5 25 0.12 1.2 3.6
0.21 0.73 0.77 7.7 23 0.11 1.1 3.3
0.19 0.66 0.71 7.1 21 0.10 1.0 3.0 Table 5 provides the cumulative amount of ketamine that permeates human skin in the Franz Diffusion Cell model disclosed in Figure 8, as provided by a transdermal delivery device according to Example 2 of the present invention. The total amount of drug in the transdermal delivery device of Example 2 is 4.75 mg. Therefore, the transdermal bioavailability of Example 2 within 24 hours is about 78%.
TABLE 5
Figure imgf000018_0001
Transdermal delivery devices can be abused. One method of abuse is to place the device in a solvent to separate the drug from the polymeric matrix, followed by separating the drug from any additional components. In order to deter abuse, the present invention is further directed towards a novel transdermal delivery device comprising ketamine and abuse deterrent agents.
Abuse deterrent agents are employed because they have one or more of the following properties: (1) unpalatable bitterness or other repulsive tastes in the mouth (i.e., bittering agents); (2) formation of gel upon mixing with the extraction solvents (i.e., gel forming agents); (3) severe irritation when injected (i.e. , irritants); (4) mood depression (e.g., droperidol) or other pronounced central nervous system (CNS) effects; (5) acute gastrointestinal, cardiac or respiratory effects; (6) violent nausea or vomiting; (7) repugnant smells if not used as instructed; (8) sleep inducing, thereby causing the abuser to miss or be made unaware of the euphoria; and/or (9) deactivation or degradation of the active ingredient (i.e., strong oxidation agents (such as hydrogen peroxide), strong acid, or strong base, and/or antagonists) upon attempted extraction. The abuse deterrent agent is employed at about 0.01-10% by weight of the final formulation, preferably about 0.1-4% by weight, and most preferably about 0.1-0.5% by weight.
The abuse deterrent agents can be included in the adhesive drug matrix or in a separate abuse deterrent layer (also referred to as the abuse deterrent matrix). The abuse deterrent layer may be comprised of a combination of polymer and the abuse deterrent agent. Additionally, the abuse deterrent layer can be the abuse deterrent agent itself because many of the recited polymers also act as gel forming agents. Suitable polymers include, but are not limited to, one or more pharmaceutically acceptable polymers that will undergo an increase in viscosity upon contact with a solvent. Preferred polymers include polyethylene oxide, polyvinyl alcohol, hydroxypropyl methylcellulose, carbomers (carbopol), polyvinylpyrrolidone (PVP), and/or other cellulose polymers. In one embodiment of the present invention the polymer includes polyethylene oxide. The polyethylene oxide can have an average molecular weight ranging from about 300,000-5,000,000, and more preferably from about 600,000-5,000,000, and most preferably at least about 5,000,000. In one embodiment, the polyethylene oxide is a high molecular weight polyethylene oxide. Examples of suitable, commercially available polyethylene oxide polymers include Poly ox®, WSRN-1105 and/or WSR coagulant, available from Dow Chemical. The preferred weight range of the polymer is from about 1-40% by weight of the final formulation, and the most preferred range of the polymer is from about 2- 10% by weight of the final formulation.
Bittering agents are pharmaceutically acceptable bitter substances that create a bitter taste or effect when administered nasally (snorted), orally, buccally or sublingually, making consumption difficult. The bittering agents that may be employed in the present invention include, but are not limited to, sucrose octaacetate (used as a denaturant for alcohol) (e.g., SD- 40), denatonium saccharide, denatonium benzoate, caffeine, quinine (or a quinine salt such as quinine sulfate), bitter orange peel oil, and other botanical extract ingredients, such as pepper extract (cubeb), capsicum, and the like. Preferred bittering agents are sucrose octaacetate, denatonium benzoate (Bitrex), and denatonium saccharide (four times more bitter than denatonium benzoate) because they are extremely bitter even at low concentrations and are essentially non-toxic. The bittering agent is employed at about 0.01-10% by weight of the final formulation, preferably about 0.1-4% by weight, and most preferably about 0.1-0.5% by weight.
Gel forming agents are employed to form a gel structure upon mixing with the extraction solvents and, thus, provide abuse deterrent properties. Specifically, gel forming agents are compounds that upon contact with a solvent (e.g., water or alcohol), absorb the solvent and swell, thereby forming a viscous or semi-viscous substance that significantly reduces and/or minimizes the amount of free solvent which can contain an amount of solubilized drug, and which minimizes what can be drawn into a syringe for injection (i.e. , IV or intramuscular). The gel can also reduce the overall amount of drug extractable with the solvent by entrapping the drug in a gel matrix. In certain embodiments the gel forming agent can be in a segregated abuse deterrent layer laminated to the adhesive drug matrix.
The gel forming agents that may be employed include, but are not limited to, ethyl cellulose, cellulose acetate, cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, cellulose ether, cellulose ester, cellulose ester ether, acrylic resins comprising copolymers synthesized from acrylic and methacrylic acid esters, the acrylic polymer may be selected from the group consisting of acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethylmethacrylates, cyanoethyl methacrylate, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, poly(methacrylic acid anhydride), glycidyl methacrylate copolymers, and mixtures thereof. The gel forming agent is preferably employed at about 3-40% by weight of the final formulation, and the most preferably about 5-20% by weight of the final formulation.
In embodiments of the present invention the gel forming agent includes polyvinyl alcohol with a molecular weight ranging from about 20,000-200,000, specific gravity ranging from about 1.19-1.31, and viscosity ranging from about 4-65 cps. The polyvinyl alcohol used in the formulation is preferably a water-soluble synthetic polymer represented by — (— C2H4 O— )n— , where n can range from about 500-5,000. Examples of suitable, commercially available polyvinyl alcohol polymers include PVA, USP, available from Spectrum Chemical Manufacturing Corporation, New Brunswick, N.J. 08901.
In embodiments of the present invention, the gel forming agent includes hydroxypropyl methyl cellulose (Hypromellose) with a molecular weight ranging from about 10,000- 1,500,000, typically from about 5000-10,000 (i.e., low molecular). The specific gravity of the hydroxypropyl methyl cellulose ranges from about 1.19-1.31, with an average specific gravity of about 1.26. Viscosity of the hydroxypropyl methyl cellulose is about 3600-5600 cPs. The hydroxypropyl methylcellulose used in the formulation can be a water-soluble synthetic polymer. Examples of suitable, commercially available hydroxypropyl methylcellulose polymers include Methocel K100 LV and Methocel K4M, available from Dow chemicals.
In other embodiments of the present invention the gel forming agent includes hydrophilic polymers, such as hydrogels, which provides viscosity to the dosage form upon tampering. In such embodiments, when an abuser crushes and dissolves the dosage form in a solvent (e.g., water or saline), a viscous or semi-viscous gel is formed.
In certain embodiments of the present invention, the gel forming agent can include carbomers, having a molecular weight ranging from 700,000-4,000,000 and viscosity ranging from about 4000-39,400 cPs. Carbomer is preferably employed in the present invention from about 1-40% by weight of the final formulation, and most preferably from about 2-10% by weight. Examples of suitable, commercially available carbomers include carbopol 934P NF, carbopol 974P NF, and carbopol 97 IP NF, available from Lubrizol.
Irritants are pharmaceutically inert compounds that induce irritation to the mucous membranes of the body (i.e. , nasal, mouth, eye, intestine, urinary tract). The irritants that may be employed in the present invention include, but are not limited to surfactants, such as sodium lauryl sulfate (SLS), poloxamer, sorbitan monoesters and glyceryl monooleates, as well as spicy ingredients, and others. The irritants are preferably employed at about 0.01-10% by weight of the final formulation, preferably 0.01-10% by weight, and most preferably about 0.1- 5% by weight.
In embodiments of the present invention, the irritant can deter abuse upon tampering with the transdermal delivery device. For example, if an abuser extracts and dries the ketamine, then the irritant is exposed and discourages inhalation of the ketamine mixed with the irritant, as inhalation (e.g., via snorting through the nose) will induce pain and/or irritation of the abuser's mucous membrane and/or nasal passageway tissue.
Other suitable excipients useful in the preparation of transdermal delivery devices are within the knowledge of those skilled in the art, and can be found in the Handbook of Pharmaceutical Excipients (7th ed. 2012), the entire content of which is hereby incorporated by reference.
EXAMPLES
Examples 1-5
The formulation of Examples 1-5 are disclosed below in Table 6.
TABLE 6
Figure imgf000022_0001
The pressure sensitive adhesive (PSA) employed in Examples 1, 2, and 4 was DuroTak 387-2052 (supplied by Henkel Adhesives). The PSA employed in Example 3 was DuroTak 87-2677 (Henkel Adhesives). The PSA employed in Example 5 was Duro-Tak 87-4098 (Henkel Adhesives). Persons skilled in the art will understand that other known pressure sensitive adhesives can be readily employed with the transdermal delivery devices of the present invention.
The skin permeation enhancer employed in Examples 4 and 5 was diethylene glycol monoethyl ether, sold under the tradename Transcutol P. Persons skilled in the art will understand that other known skin permeation enhancers can be readily employed with the transdermal delivery devices of the present invention.
The crystallization inhibitor in Examples 3 and 4 was polyvinyl pyrrolidone-co-vinyl acetate, sold under the tradename Kollidon VA 64 (BASF Corporation). The crystallization inhibitor employed in Example 5 was polymathacrylate-based polymer, sold under the tradename Plastoid B (Evonik Corporation). Persons skilled in the art will understand that other known crystallization inhibitor enhancers can be readily employed with the transdermal delivery devices of the present invention.
Figure 5 is a comparison of the plasma concentration-time profile of a 0.5 mg/kg dose of ketamine in a human subject following: (1) a 40-minute single IV administration; and (2) administration of a 24 hour transdermal delivery device of the present invention according Example 2 (a 9.4 cm2 transdermal delivery device with a 3.75 mg/cm2 permeation rate). Convolution analysis was applied in accordance with the pharmacokinetic parameters set forth in Fanta, et al., Eur. J. Clin. Pharmacol, 71 :441-447 (2015). The transdermal delivery device according to Example 2 exhibits a lower Cmax, preferably less than about 30%, and more preferably less than about 20%, of the Cmax from an equivalent IV dosage.
Figure 6 discloses ketamine plasma concentration-time profiles for three sizes (10, 100 and 300 cm2) of the once-a-day transdermal delivery device of the present invention according to Example 2.
Figure 7 discloses ketamine plasma concentration-time profiles for three sizes (10, 100 and 300 cm2) of the three times-a-day transdermal delivery device of the present invention according to Example 2.
Skin permeation enhancers are incorporated in the transdermal delivery devices of the present invention to ensure that sufficient ketamine can penetrate through skin. Skin permeation studies were performed on the transdermal delivery devices prepared according to Examples 1-5 using Franz diffusion cells maintained at 37° C. The receptor medium was phosphate buffered saline at pH 7.4, the receptor volume was 12 ml and the permeation area was 1.767 cm2. Human cadaver skin was used and the tests were performed in triplicate. A lxl inch transdermal delivery device was placed onto the donor side of the skin diffusion cells, adhered onto the skin, and the experiment was initiated with the receptor medium being continuously mixed (stirring at 600 rpm). Samples (1.5 ml) of the receptor phase were obtained at 2, 4, 8, 12, 24, 48 and 72 hours. The drug concentrations were quantitated using HPLC. As demonstrated in Table 6, Examples 1-5 of the present invention all provide good skin permeability. The cumulative amounts of ketamine that permeated after 24 hours are shown in Table 7. TABLE 7
Figure imgf000024_0001
Figure 8 depicts in vitro skin permeability of the transdermal delivery devices of the present invention according to Example 2 as shown by the Franz Diffusion Cell model.
Drug crystallization will retard drug release and skin permeability, reducing the efficacy of the transdermal delivery device. Drug crystals should not be formed in the transdermal delivery device over a period approximating the shelf life, i.e., for about 6 months or greater. Examples 1 and 2 showed instability, i.e. drug crystals were formed in the adhesive drug matrix 4-7 days after preparation of the transdermal delivery devices. Examples 3, 4 and 5, were found to be stable for at least 4 weeks, i.e. within this period, no crystals were formed. Example 5 was found to be stable for at least 6 months at conditions of 25° C, 60% RH.
Table 8 reports the stability data for the transdermal delivery devices prepared according to Examples 1-5.
Figure 9 depicts the stability of ketamine in a transdermal delivery device according to Example 5 after 0 months and after 6 months.
TABLE 8
Figure imgf000024_0002
Example 6:
Transdermal Delivery Device With Non-segregated Abuse Deterrent Agents
Example 5 was modified to prepare a transdermal delivery device that contains 5 mg of denatonium benzoate and 200 mg of ketamine in the adhesive drug matrix. In vitro skin permeation studies showed that no denatonium benzoate permeated the skin (because of its large molecular weight (447 DA) and the high melting point (170° C). Nevertheless, ketamine showed excellent skin permeability (0.8 mg/cm2 in 24 hours), indicating that incorporation of an abuse deterrent agent, such as denatonium benzoate did not affect the skin permeation of ketamine.
In an extraction study to simulate attempted drug-abuse, transdermal delivery devices prepared according to Example 6 were soaked for 60 minutes in 100 ml of three different media: (1) 40% ethanol; (2) 70% isopropyl alcohol; and (3) acetone. All three media were assayed for ketamine and denatonium benzoate using HPLC. More than 50% of the original ketamine and more than 50% of the denatonium benzoate were found in the media (i.e. , the bittering agent extracted proportionally to the amount of ketamine extracted), indicating the effectiveness of using denatonium benzoate as an abuse deterrent agent in the present invention.
Example 7:
Transdermal Delivery Device with Abuse Deterrent Agents in a Segregated Layer
Example 7 comprises a transdermal delivery device with an abuse deterrent agent in a segregated layer. The abuse deterrent agent employed in Example 7 is a gelling agent which reacts with common solvents (e.g., water and alcohol) used to extract and abuse the ketamine in the transdermal delivery device. The thickness of the adhesive-drug layer and the abuse deterrent layer are both about 2 to about 5 mils. The transdermal delivery device according to Example 7 is prepared in a two-step process.
Step 1. Preparation of the Abuse Deterrent Layer
Poly Ox 1105, propylene glycol, and PEG 4000 were mixed to form the abuse deterrent layer. The three ingredients were dissolved in a water/ethanol solvent, followed by casting of the wet film directly on a sheet of backing layer, e.g., 3M's polyethylene film, Scotpak 1012. The wet film is then dried at 60° C for 30 min in a convective-air drying oven. The coating thickness of the abuse deterrent layer is about 3 mils. An example of a suitable abuse deterrent layer composition is disclosed in Table 9. TABLE 9
Figure imgf000026_0001
evaporated during processing.
An abuse deterrent agent, such as Bitrex and sodium lauryl sulfate (SLS), may be incorporated into the abuse deterrent layer, preferably from about 0.01-5% by weight of the final formulation, and most preferably from about 0.05-0.5% by weight of the final formulation.
Step 2. Preparation of Adhesive Drug Matrix Layer:
The adhesive drug matrix layer is prepared by casting the adhesive drug matrix mix directly on the abuse deterrent layer (prepared in step 1), or onto a release liner and then laminated to the abuse deterrent layer.
DuroTak 87-4098 is weighed into a 100 ml beaker, and then mixed at low speed. Next, the Kollidon VA 64 and the ketamine are added to the mixer. The batch is mixed until all ingredients are dissolved. Then wet films are prepared at 3 mils thickness using a film casting applicator on release liner, such as 3M's 9744. The wet coating is air dried for 1 hour, and then oven dried at 60° C for 10 min. Finally, the laminate is dried onto the abuse deterrent layer, which was subsequently coated on 3M's backing Scotpak 1012.
The laminated sheet can be die-cut into transdermal delivery devices of various sizes, such as 10 cm2, 20 cm2, 100 cm2, 300 cm2 to obtain the desired dosages of the drug.
An exemplary composition of the adhesive drug matrix is given in Table 10. TABLE 10
Figure imgf000027_0001
Example 8
A transdermal delivery device according to Examples 7 is prepared with apomorphine, an emetic, in the abuse deterrent layer. After solvent extraction by an abuser, the emetic can cause severe nausea if injected, snorted, or inhaled. Apomorphine is preferably employed in the present invention from about 0.05-5% by weight of the final formulation, and most preferred from about 0.1-2% by weight.
Example 9
Example 9 was prepared in accordance with Example 7, with the exception that the abuse deterrent agent is capsaicin. After being dissolved in solvents by an abuser, the ketamine solution containing capsaicin will cause a torturous buming sensation if snorted or inhaled, thereby reducing the abuse potential of the transdermal drug delivery device.
Examples 10
Table 11 provides additional techniques by which the abuse deterrent agents can be employed in the transdermal delivery device of the present invention.
TABLE 11
Figure imgf000027_0002
Example 11
In an embodiment of the present invention is prepared according to Example 7, in which the adhesive-drug layer comprises an adhesive and 200 mg of ketamine, and the abuse deterrent layer comprises a gel forming agent comprising 7 mg of SLS and 5 mg of denatonium benzoate.
The foregoing description and examples have been set forth merely to illustrate the present invention and are not intended to be limiting. Since modifications of the described embodiments incorporating the spirit and substance of the invention may occur to persons skilled in the art, the invention should be construed broadly to include all variations within the scope of this application, including but not limited to the appended claims and equivalents thereof.

Claims

Claims
1. A transdermal delivery device comprising ketamine.
2. The transdermal delivery device according to claim 1, further comprising a pressure sensitive adhesive, a skin permeation enhancer, and a crystallization inhibitor.
3. The transdermal delivery device according to claim 2, comprising about 1 -35 weight percent of ketamine, about 30-90 weight percent of pressure sensitive adhesive, about 1-10 weight percent of skin permeation enhancer, and about 5-40 weight percent of crystallization inhibitor.
4. The transdermal delivery device according to claim 1, further comprising about 0.01- 10 weight percent of abuse deterrent agent.
5. The transdermal delivery device according to claim 4, wherein the abuse deterrent agent is selected from capsaicin, apomorphine, denatonium, sodium laurel sulfate, a gel forming agent, and combinations thereof.
6. A method of treating major depressive disorders comprising administration of a transdermal delivery device comprising ketamine.
7. The method according to claim 6 wherein the transdermal delivery device is suitable for administration for about 8 hours to about 7 days.
8. A method of treating pain comprising administration of a transdermal delivery device comprising ketamine.
9. The method according to claim 8 wherein the transdermal delivery device is suitable for administration for about 8 hour to about 7 days.
10. The transdermal delivery device according to claim 1, which provides about 0.1-30 mg/day/cm2 of ketamine for about 7 days.
1 1. The transdermal delivery device according to claim 1, which provides about 0.1-30 mg/day/cm2 of ketamine for about 1 day.
12. The transdermal delivery device according to claim 1, which provides about 0.1-30 mg/day/cm2 of ketamine for about 3.5 days.
13. The transdermal delivery device according to claim 1, which provides about 0.5-5 mg/day/cm2 of ketamine for about 7 days.
14. The transdermal delivery device according to claim 1, which provides about 0.5-5 mg/day/cm2 of ketamine for about 1 day.
15. The transdermal delivery device according to claim 1, which provides about 0.5-5 mg/day/cm2 of ketamine for about 3.5 days.
16. The transdermal delivery device according to claim 1, wherein said device is about 10-300 cm2.
17. The transdermal delivery device according to claim 16, wherein said device is about 100-300 cm2.
18. The transdermal delivery device according to claim 16, wherein said device is about 10-100 cm2.
19. The transdermal delivery device according to claim 1, wherein the transdermal delivery device provides reduced Cmax compared to immediate release administration of ketamine.
20. The transdermal delivery device according to claim 1, which provides ketamine plasma concentrations from about 0.4-4000 ng/ml for about 8-168 hours.
21. The transdermal delivery device according to claim 20 which provides ketamine plasma concentrations from about 10-200 ng/ml for treatment of major depressive disorders.
22. The transdermal delivery device according to claim 20 which provides ketamine plasma concentrations below 100 ng/ml for about 8 hours to about 7 days, and which reduces the adverse side effect of ketamine when compared with plasma concentrations from immediate release ketamine formulations.
23. The transdermal delivery device according to claim 20 which provides ketamine plasma concentrations from about 50-1000 ng/ml for treatment of pain.
24. The transdermal delivery device according to claims 20 which provides reduced plasma fluctuations over time when compared with plasma concentrations from immediate release ketamine formulations.
25. The transdermal delivery device according to claim 1, which provides:
(a) reduced ^max fluctuations,
(b) reduced plasma concentration fluctuations, and
(c) reduced adverse side effects, when compared with ketamine plasma concentrations from immediate release formulations.
26. The transdermal delivery device according to claim 1, wherein the Cmax is no greater than about 30% of the Cmax from a dose equivalent immediate release ketamine formulation.
27. The transdermal delivery device according to claim 1, wherein said ketamine is the R- enantiomer.
28. The transdermal delivery device according to claim 1, wherein said ketamine is the S- enantiomer.
PCT/US2016/039601 2015-06-27 2016-06-27 Ketamine transdermal delivery system WO2017003935A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2017566151A JP6845162B2 (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
KR1020187002315A KR102299359B1 (en) 2015-06-27 2016-06-27 Ketamine Transdermal Delivery System
AU2016288188A AU2016288188B2 (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
CN201680037543.8A CN107847469A (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
US15/736,182 US11191734B2 (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
EP16818548.6A EP3297620A4 (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
CA2987909A CA2987909C (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
HK18108421.2A HK1248578A1 (en) 2015-06-27 2018-06-29 Ketamine transdermal delivery system
US17/540,695 US20220193000A1 (en) 2015-06-27 2021-12-02 Ketamine transdermal delivery system

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562185573P 2015-06-27 2015-06-27
US62/185,573 2015-06-27

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/736,182 A-371-Of-International US11191734B2 (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system
US17/540,695 Continuation US20220193000A1 (en) 2015-06-27 2021-12-02 Ketamine transdermal delivery system

Publications (1)

Publication Number Publication Date
WO2017003935A1 true WO2017003935A1 (en) 2017-01-05

Family

ID=57609546

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/039601 WO2017003935A1 (en) 2015-06-27 2016-06-27 Ketamine transdermal delivery system

Country Status (9)

Country Link
US (2) US11191734B2 (en)
EP (1) EP3297620A4 (en)
JP (1) JP6845162B2 (en)
KR (1) KR102299359B1 (en)
CN (1) CN107847469A (en)
AU (1) AU2016288188B2 (en)
CA (1) CA2987909C (en)
HK (1) HK1248578A1 (en)
WO (1) WO2017003935A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107823195A (en) * 2017-11-24 2018-03-23 无锡市精神卫生中心 Application of the R ketamines in depression acute stages treated
WO2018195318A1 (en) * 2017-04-20 2018-10-25 Guangzhou Dazhou Biomedicine Ltd. Transdermal drug delivery system for ketamine
JP2018188420A (en) * 2017-04-28 2018-11-29 日東電工株式会社 Transdermal absorption preparation
WO2020086673A1 (en) * 2018-10-26 2020-04-30 Guangzhou Dazhou Biomedicine Ltd. Ketamine oral transmucosal delivery system
EP3725306A1 (en) 2019-04-17 2020-10-21 LTS Lohmann Therapie-Systeme AG Transdermal therapeutic system
WO2020212596A1 (en) 2019-04-17 2020-10-22 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system
US10869844B2 (en) 2014-09-15 2020-12-22 Janssen Pharmaceutica Nv Methods for the treatment of depression
WO2021037960A1 (en) * 2019-08-30 2021-03-04 Klaria Pharma Holding Ab Pharmaceutical formulation
JP2021522240A (en) * 2018-04-25 2021-08-30 シンケイ セラピューティクス リミテッド ライアビリティ カンパニー Tetrabenazine transdermal delivery device
US11219600B2 (en) 2017-06-08 2022-01-11 Klaria Pharma Holding Ab Pharmaceutical formulation
EP3949956A1 (en) * 2020-08-06 2022-02-09 LTS Lohmann Therapie-Systeme AG Esketamine-suspension-tts
US11446260B2 (en) 2013-03-15 2022-09-20 Janssen Pharmaceutica Nv Pharmaceutical composition of S-ketamine hydrochloride
US11491120B2 (en) 2017-11-09 2022-11-08 The Trustees Of Columbia University In The City Of New York Pharmacological prophylactics against stress-induced affective disorders in females
US11559497B2 (en) 2020-06-19 2023-01-24 Guangzhou Dazhou Biomedicine Ltd. Transdermal drug delivery system for ketamine
RU2791107C1 (en) * 2019-04-17 2023-03-02 Лтс Ломанн Терапи-Системе Аг Transdermal therapeutic system
US11622948B2 (en) 2017-11-09 2023-04-11 The Trustees Of Columbia University In The City Of New York Biomarkers for efficacy of prophylactic treatments against stress-induced affective disorders
US11707440B2 (en) 2017-12-22 2023-07-25 Janssen Pharmaceuticals, Inc. Esketamine for the treatment of depression
US11752114B2 (en) 2019-04-17 2023-09-12 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system
US11883526B2 (en) 2019-03-05 2024-01-30 Janssen Pharmaceutica Nv Esketamine for the treatment of depression

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019213551A1 (en) 2018-05-04 2019-11-07 Perception Neuroscience, Inc. Methods of treating substance abuse
CN111346073B (en) * 2018-12-21 2022-06-10 宜昌人福药业有限责任公司 Transdermal drug delivery pharmaceutical composition and preparation method and application thereof
CN109966272A (en) * 2019-04-30 2019-07-05 深圳市泛谷药业股份有限公司 A kind of ketamine transdermal patch and preparation method thereof
CN117797130A (en) * 2022-09-30 2024-04-02 宜昌人福药业有限责任公司 S-ketamine percutaneous composition and preparation method and application thereof
WO2024067729A1 (en) * 2022-09-30 2024-04-04 宜昌人福药业有限责任公司 S-ketamine transdermal composition and preparation method therefor and use thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110065627A1 (en) * 2008-11-21 2011-03-17 Cymbiotics Inc. Transdermal delivery of medicaments with combinations of cetylated fatty fatty ester penetrant complexes
US20110178114A1 (en) * 2008-06-30 2011-07-21 Afgin Phara Llc Topical regional neuro-affective therapy
US20120270916A1 (en) * 2002-11-25 2012-10-25 Taraxos Inc. Topical formulations for treating neuropathy
US20140256821A1 (en) * 2006-03-22 2014-09-11 Icahn School Of Medicine At Mount Sinai Intrannasal administration of ketamine to treat depression

Family Cites Families (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5635204A (en) * 1994-03-04 1997-06-03 Montefiore Medical Center Method for transdermal induction of anesthesia, analgesia or sedation
CA2230690C (en) * 1995-08-30 2008-12-23 Stuart L. Weg Administration of ketamine to manage pain and to reduce drug dependency
US6248789B1 (en) 1996-08-29 2001-06-19 Stuart L. Weg Administration of ketamine to manage pain and to reduce drug dependency
US6017961A (en) 1999-07-08 2000-01-25 Flores; John Anthony Ketamine and n-butyl-p-aminobezoate in PLO
EP1395289B1 (en) 2001-06-07 2010-12-15 Christine Dr. Sang Treatment of neuropathic pain with a n-methyl-d-aspartate (nmda) receptor antagonists
US20030082225A1 (en) * 2001-10-19 2003-05-01 Mason Paul Arthur Sterile, breathable patch for treating wound pain
US6855735B2 (en) 2002-03-20 2005-02-15 Temple University Of The Commonwealth System Of Higher Education Ketamine treatment of restless legs syndrome
US20050148673A1 (en) 2002-07-11 2005-07-07 Harbut Ronald E. Prolonged administration of NMDA antagonist and safener drug to alter neuropathic pain condition
EP1551393A4 (en) 2002-07-30 2010-06-16 Peter Migaly Combination therapy for depression, prevention of suicide, and varous medical and psychiatric conditions
US7273889B2 (en) 2002-09-25 2007-09-25 Innovative Drug Delivery Systems, Inc. NMDA receptor antagonist formulation with reduced neurotoxicity
EP1567145B1 (en) 2002-11-18 2009-06-03 Yaupon Therapeutics, Inc. Analgesic uses of (s)-norketamine
US10172810B2 (en) 2003-02-24 2019-01-08 Pharmaceutical Productions, Inc. Transmucosal ketamine delivery composition
US20040265364A1 (en) 2003-06-25 2004-12-30 Binnur Ozturk Neuropathy cream
US20050222270A1 (en) 2004-02-26 2005-10-06 Olney John W Prolonged administration of NMDA antagonist drug and safener drug to create improved stable neural homeostasis
US8741332B2 (en) 2004-06-07 2014-06-03 Nuvo Research Inc. Compositions and methods for dermally treating neuropathic pain
WO2006096518A2 (en) 2005-03-04 2006-09-14 Neurosystec Corporation Improved gacyclidine formulations
US7645767B2 (en) 2006-08-31 2010-01-12 Trinity Laboratories, Inc. Pharmaceutical compositions for treating chronic pain and pain associated with neuropathy
JP2010525081A (en) 2007-04-26 2010-07-22 オースペックス・ファーマシューティカルズ・インコーポレイテッド Deuterium labeled ketamine
AU2008260774B2 (en) * 2007-06-08 2011-01-06 Samyang Biopharmaceuticals Corporation Matrix-type transdermal drug delivery system and preparation method thereof
US8629184B2 (en) 2008-09-27 2014-01-14 TARAXOS, Inc. Topical formulations for treatment of neuropathy
JP2010229045A (en) 2009-03-26 2010-10-14 Mikasa Seiyaku Co Ltd Percutaneous absorption type preparation
US20120225949A1 (en) 2009-08-14 2012-09-06 Demitri Papalos Compositions and methods for treating bipolar disorder
KR101292768B1 (en) * 2010-04-23 2013-08-05 아이큐어 주식회사 Transdermal Drug Delivery System
SI2582366T1 (en) 2010-06-15 2016-02-29 Gruenenthal Gmbh Pharmaceutical combination for the treatment of pain
DE102010024105A1 (en) * 2010-06-17 2011-12-22 Grünenthal GmbH Transdermal administration of memantine
WO2013003669A2 (en) 2011-06-30 2013-01-03 University Of South Florida Compositions, methods of use, and methods of treatment
US9724315B2 (en) * 2011-09-30 2017-08-08 Cmpd Licensing, Llc Compounded transdermal pain management
CN109793725B (en) 2011-10-14 2023-07-25 美国政府健康及人类服务部 Pharmaceutical composition for treating melancholia and neuropathic pain and application thereof
WO2013088135A1 (en) 2011-12-14 2013-06-20 Astrazeneca Ab Gabr-a2 diagnostic
EP2809307B1 (en) * 2012-01-31 2017-12-20 Grünenthal GmbH Pharmaceutical patch for transdermal administration of (1r,4r)-6'-fluoro-n,n-dimethyl-4-phenyl-4',9'-dihydro-3'h-spiro[cyclohexane-1,1'-pyrano[3,4-b]indol]-4-amine
US20130209585A1 (en) 2012-02-13 2013-08-15 Stanley Kim Topical NMDA Antagonist Formulations for the Treatment of Peripheral Neuropathy
US20130236573A1 (en) 2012-03-12 2013-09-12 Janssen Pharmaceutica Nv Esketamine for the treatment of treatment-refractory or treatment-resistant depression
JP2015512418A (en) 2012-03-30 2015-04-27 ザ ジェネラル ホスピタル コーポレイション Compositions containing scopolamine and ketamine in the treatment of depression
US20140079740A1 (en) 2012-08-02 2014-03-20 ClinPharm Support GmbH Oral transmucosal adminstration forms of s-ketamine
JP6722453B2 (en) 2012-08-23 2020-07-15 スチュアート エル ウェグ Anxiolytic composition, formulation and method of use
FR2994844B1 (en) 2012-08-31 2015-01-02 Assist Publ Hopitaux De Paris GELIFYING FORMULATION BASED ON KETAMINE
JP6309957B2 (en) 2012-10-08 2018-04-11 オークランド ユニサービシーズ リミティド Ketamine derivatives
EP3636251B1 (en) 2013-03-15 2022-01-19 Janssen Pharmaceutica NV Pharmaceutical composition of s-ketamine hydrochloride
KR20220044380A (en) 2013-04-12 2022-04-07 이칸 스쿨 오브 메디슨 엣 마운트 시나이 Method for treating post-traumatic stress disorder
US9913803B2 (en) 2013-08-26 2018-03-13 Amorsa Therapeutics, Inc. Single-layer oral dose of neuro-attenuating ketamine
RS62516B1 (en) 2013-09-13 2021-11-30 Univ Chiba Nat Univ Corp Application of r-ketamine and salt thereof as pharmaceuticals
WO2015051259A1 (en) 2013-10-04 2015-04-09 Impax Laboratories, Inc. Pharmaceutical compositions and methods of use
CA2923064C (en) * 2014-02-20 2016-10-25 Cura Health Inc. Transdermal composition for treating pain
US11207316B2 (en) 2014-05-30 2021-12-28 West Virginia University Ketamine or dextromethorphan formulations and methods of use
JP6545788B2 (en) 2014-08-13 2019-07-17 ヤンセン ファーマシューティカ エヌ.ベー. How to treat depression
FI3215147T3 (en) 2014-11-04 2024-04-18 Acadia Pharm Inc Neuro-attenuating norketamine compounds and methods
US20180015054A1 (en) 2014-12-31 2018-01-18 Icahn School Of Medicine At Mount Sinai Method of Maintaining the Anti-Depressant Effect of Ketamine With Lithium
WO2016172672A1 (en) 2015-04-24 2016-10-27 Icahn School Of Medicine At Mount Sinai Method for treating suicidal ideation
US20160338977A1 (en) 2015-05-20 2016-11-24 Janssen Pharmaceutica Nv Methods and kits for treating depression
WO2017041112A1 (en) 2015-09-04 2017-03-09 Cyprus Therapeutics, Inc. Ketamine and cytochrome p 450 inhibitor combinations
WO2017087691A1 (en) 2015-11-17 2017-05-26 The Trustees Of Columbia University In The City Of New York Pharmacological prophylactics against stress-induced affective disorders and their associated symptoms
WO2017139382A1 (en) 2016-02-08 2017-08-17 The Texas A&M University System Combination of adjuvant drugs esketamine and brimonidine for medical treatments
WO2017139768A1 (en) 2016-02-12 2017-08-17 Synergistic Therapeutics, Llc Sublingual antidepressant lozenge
US20170266319A1 (en) 2016-03-21 2017-09-21 Children's Hospital Medical Center Methods and compositions for treating conditions associated with spinal cord injury
US20190083420A1 (en) 2016-03-25 2019-03-21 The Uinited States of America, as represented by the Secretary, Department of Health and Human Serv Methods of using (2r, 6r)-hydroxynorketamine and (2s, 6s)-hydroxynorketamine in the treatment of depression, anxiety, anhedonia, fatigue, suicidal ideation, and post traumatic stress disorders
EP3433230A1 (en) 2016-03-25 2019-01-30 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Crystal forms and methods of synthesis of (2r, 6r)-hydroxynorketamine and (2s, 6s)-hydroxynorketamine
CA3019565A1 (en) 2016-04-01 2017-10-05 Good Pharmaceutical Development Company, Llc Topical compositions for neuropathic pain
CN108884019A (en) 2016-04-11 2018-11-23 克雷西奥生物科技有限公司 Deuterated chloramines ketone derivatives
US20170348252A1 (en) 2016-04-21 2017-12-07 Robert Kent Preparation and Use of Ketamine Derivatives and Ketamine Modifiers in the Treatment of Pain
CN109475500A (en) 2016-05-25 2019-03-15 财团法人卫生研究院 The method and composition of psychiatric symptoms side effect and habituation illness are caused for lowering ketamine
WO2017208031A1 (en) 2016-06-03 2017-12-07 Small Pharma Ltd Solid oral dosage forms of 2r,6r-hydroxynorketamine or derivatives thereof
US20200030251A1 (en) 2017-04-20 2020-01-30 Guangzhou Dazhou Biomedicine Ltd. Transdermal drug delivery system for ketamine

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120270916A1 (en) * 2002-11-25 2012-10-25 Taraxos Inc. Topical formulations for treating neuropathy
US20140256821A1 (en) * 2006-03-22 2014-09-11 Icahn School Of Medicine At Mount Sinai Intrannasal administration of ketamine to treat depression
US20110178114A1 (en) * 2008-06-30 2011-07-21 Afgin Phara Llc Topical regional neuro-affective therapy
US20110065627A1 (en) * 2008-11-21 2011-03-17 Cymbiotics Inc. Transdermal delivery of medicaments with combinations of cetylated fatty fatty ester penetrant complexes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3297620A4 *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11446260B2 (en) 2013-03-15 2022-09-20 Janssen Pharmaceutica Nv Pharmaceutical composition of S-ketamine hydrochloride
US11173134B2 (en) 2014-09-15 2021-11-16 Janssen Pharmaceutica Nv Methods for the treatment of depression
US11311500B2 (en) 2014-09-15 2022-04-26 Janssen Pharmaceutica Nv Methods for the treatment of depression
US10869844B2 (en) 2014-09-15 2020-12-22 Janssen Pharmaceutica Nv Methods for the treatment of depression
WO2018195318A1 (en) * 2017-04-20 2018-10-25 Guangzhou Dazhou Biomedicine Ltd. Transdermal drug delivery system for ketamine
CN110913844A (en) * 2017-04-20 2020-03-24 广州大洲生物医药科技有限公司 Transdermal drug delivery system for ketamine
JP2018188420A (en) * 2017-04-28 2018-11-29 日東電工株式会社 Transdermal absorption preparation
US11179469B2 (en) 2017-04-28 2021-11-23 Nitto Denko Corporation Transdermal absorption preparation
US11904049B2 (en) 2017-06-08 2024-02-20 Klaria Pharma Holding Ab Pharmaceutical formulation
US11219600B2 (en) 2017-06-08 2022-01-11 Klaria Pharma Holding Ab Pharmaceutical formulation
US11622948B2 (en) 2017-11-09 2023-04-11 The Trustees Of Columbia University In The City Of New York Biomarkers for efficacy of prophylactic treatments against stress-induced affective disorders
US11491120B2 (en) 2017-11-09 2022-11-08 The Trustees Of Columbia University In The City Of New York Pharmacological prophylactics against stress-induced affective disorders in females
CN107823195A (en) * 2017-11-24 2018-03-23 无锡市精神卫生中心 Application of the R ketamines in depression acute stages treated
US11707440B2 (en) 2017-12-22 2023-07-25 Janssen Pharmaceuticals, Inc. Esketamine for the treatment of depression
JP2021522240A (en) * 2018-04-25 2021-08-30 シンケイ セラピューティクス リミテッド ライアビリティ カンパニー Tetrabenazine transdermal delivery device
JP7386544B2 (en) 2018-04-25 2023-11-27 シンケイ セラピューティクス インコーポレイテッド Tetrabenazine transdermal delivery device
WO2020086673A1 (en) * 2018-10-26 2020-04-30 Guangzhou Dazhou Biomedicine Ltd. Ketamine oral transmucosal delivery system
US11883526B2 (en) 2019-03-05 2024-01-30 Janssen Pharmaceutica Nv Esketamine for the treatment of depression
WO2020212596A1 (en) 2019-04-17 2020-10-22 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system
AU2020257564B2 (en) * 2019-04-17 2023-03-02 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system
RU2791107C1 (en) * 2019-04-17 2023-03-02 Лтс Ломанн Терапи-Системе Аг Transdermal therapeutic system
US11752114B2 (en) 2019-04-17 2023-09-12 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system
EP3725306A1 (en) 2019-04-17 2020-10-21 LTS Lohmann Therapie-Systeme AG Transdermal therapeutic system
WO2021037960A1 (en) * 2019-08-30 2021-03-04 Klaria Pharma Holding Ab Pharmaceutical formulation
US11559497B2 (en) 2020-06-19 2023-01-24 Guangzhou Dazhou Biomedicine Ltd. Transdermal drug delivery system for ketamine
WO2022028913A1 (en) 2020-08-06 2022-02-10 Lts Lohmann Therapie-Systeme Ag Esketamine-suspension-tts
EP3949956A1 (en) * 2020-08-06 2022-02-09 LTS Lohmann Therapie-Systeme AG Esketamine-suspension-tts

Also Published As

Publication number Publication date
US20220193000A1 (en) 2022-06-23
EP3297620A1 (en) 2018-03-28
AU2016288188B2 (en) 2021-08-05
HK1248578A1 (en) 2018-10-19
JP2018518498A (en) 2018-07-12
CN107847469A (en) 2018-03-27
AU2016288188A1 (en) 2018-01-04
US20180353437A1 (en) 2018-12-13
KR102299359B1 (en) 2021-09-07
CA2987909C (en) 2022-04-26
JP6845162B2 (en) 2021-03-17
US11191734B2 (en) 2021-12-07
EP3297620A4 (en) 2019-01-09
CA2987909A1 (en) 2017-01-05
KR20180021127A (en) 2018-02-28

Similar Documents

Publication Publication Date Title
US20220193000A1 (en) Ketamine transdermal delivery system
US20220395469A1 (en) Transdermal drug delivery system for ketamine
JP6212104B2 (en) Abuse deterrent transdermal preparations of opiate agonists and agonist-antagonists
RU2408368C2 (en) Modified release bupropion salt preparations
KR101944367B1 (en) Abuse-resistant mucoadhesive devices for delivery of buprenorphine
US20210308040A1 (en) Ketamine oral transmucosal delivery system
EP2859892B1 (en) Patch
Gupta et al. Transdermal delivery: product and patent update
JP2022087276A (en) Memantine transdermal delivery systems
Abruzzo et al. Transdermal delivery of antipsychotics: rationale and current status
KR20220161416A (en) Transdermal Delivery of Dextromethorphan
JPWO2004024155A1 (en) Patch
JP5073124B2 (en) Noradrenergic / specific serotonergic antidepressant-containing transdermal absorption patch
JP2023506538A (en) Transdermal therapeutic system containing agomelatine
EP3725306A1 (en) Transdermal therapeutic system
JPWO2021202329A5 (en)
JP7352283B2 (en) Transdermal absorption preparation containing fentanyl citrate
US20230285323A1 (en) Esketamine-suspension-tts
Jyothika et al. Types of transdermal drug delivery systems: a literature report of the past decade
JP2010030909A (en) Transdermal absorption-type patch containing fentanyls, method for producing the same, method for inhibiting crystal deposition of fentanyls, and crystal deposition inhibitor
KR101964295B1 (en) Memantine Transdermal Delivery System Having Reduced Skin Irritation

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: 16818548

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2987909

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017566151

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2016818548

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016288188

Country of ref document: AU

Date of ref document: 20160627

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20187002315

Country of ref document: KR

Kind code of ref document: A