WO2019126270A1 - Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure - Google Patents

Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure Download PDF

Info

Publication number
WO2019126270A1
WO2019126270A1 PCT/US2018/066403 US2018066403W WO2019126270A1 WO 2019126270 A1 WO2019126270 A1 WO 2019126270A1 US 2018066403 W US2018066403 W US 2018066403W WO 2019126270 A1 WO2019126270 A1 WO 2019126270A1
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
sna
solvent
gel
gel formulation
Prior art date
Application number
PCT/US2018/066403
Other languages
French (fr)
Inventor
Valentina Mainero
Silvio Traversa
Original Assignee
Sienna Biopharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sienna Biopharmaceuticals, Inc. filed Critical Sienna Biopharmaceuticals, Inc.
Publication of WO2019126270A1 publication Critical patent/WO2019126270A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system

Definitions

  • Skin care formulations which include for example, primers, serums, lotions, ointments, gels, creams, foams and other products, are used on the skin for various purposes.
  • Formulations can contain one or more active agents, including in some embodiments, indolocarbazole compounds conjugated to polymer(s).
  • a gel formulation includes or consists essentially of, for example: a polymer conjugate of an indolocarbazole compound, a preservative/solvent, a solvent, a penetration enhancer, a pH adjusting agent, a gelling agent, and a vehicle.
  • the polymer conjugate of an indolocarbazole compound is SNA- 125 (formerly referred to as CT340; both terms are used interchangeably herein).
  • the SNA-125 may be present at about 0.05% to about 25% (w/w) of the gel formulation.
  • the SNA-125 may be present at about 0.5% to about 10% (w/w) of the gel formulation (e.g., about 0.2-2%, 0.5-2%, 2-6%, or 6- 10%, e.g., about 0.2%, 1.0%, 2.0%, or 5.0%, and overlapping ranges therein).
  • the SNA-125 may be present at about 5% (w/w) of the gel formulation.
  • Other reduced exposure compounds disclosed herein may be used with or instead of SNA-125.
  • the preservative/solvent may be one or more selected from benzoic acid, sorbic acid, boric acid, methylparaben, ethylparaben, propylparaben, butylparaben, sodium benzoate, sodium propionate, potassium sorbate, chlorobutanol, benzyl alcohol and phenyl ethyl alcohol, phenol, chlorocresol, o-phenyl phenol, benzalkonium chloride, cetyl pyridinium chloride, imidurea, thimerisal, propyl gallate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), citric acid, ascorbic acid, ascorbyl palmitate, alpha-tocopherol and disodium edetate.
  • benzoic acid sorbic acid
  • boric acid methylparaben
  • methylparaben ethylparaben
  • propylparaben propylpara
  • the preservative/solvent is benzyl alcohol.
  • the preservative/solvent may be present at about 0.1% to about 10% (w/w) (e.g., 0.1-3%, 3-6%, 6-10%, and overlapping ranges therein) of the gel formulation.
  • the preservative/solvent may be present at about 1% (w/w) of the gel formulation.
  • the solvent may be one or more selected from water, ethanol, propylene glycol, glycerin and polyethylene glycol.
  • the solvent is propylene glycol.
  • the solvent may be present at about 1% to about 30% (w/w) (e.g., 1-10%, 10-25%, 25-30%, and overlapping ranges therein) of the gel formulation. Alternatively, the solvent may be present at about 20% (w/w) of the gel formulation.
  • the penetration enhancer may be one or more selected from dimethylsulfoxide (‘DMSO’), Transcutol, oleic acid, azone, pyrrolidones, surfactants (e.g. SLS, Tween), urea, essential oils, terpenes and terpenoids, and phospholipids.
  • DMSO dimethylsulfoxide
  • the penetration enhancer is DMSO.
  • the penetration enhancer may be present at about 1% to about 30% (w/w) (e.g., 1-10%, 10-25%, 25-30%, and overlapping ranges therein) of the gel formulation. Alternatively, the penetration enhancer may be present at about 20% (w/w) of the gel formulation.
  • the pH adjusting agent may be one or more selected from trolamine, sodium hydroxide, hydrochloric acid, phosphoric acid, and triethylamine.
  • the pH adjusting agent is trolamine.
  • the pH adjusting agent may be used in an amount sufficient to adjust the final pH of the gel formulation (‘q.s.’) to greater than or equal to about 5.0.
  • the gelling agent may be one or more selected from hydroxyethyl cellulose, hydroxypropyl methylcellulose, carbopol, gelatin, sodium alginate, sodium carboxymethylcellulose, gelatin, polysaccharides and silica.
  • the gelling agent is hydroxyethyl cellulose (‘HEC’).
  • the gelling agent may be present at about 0.05% to about 20% (w/w) (e.g., 0.05-5%, 6-15%, 15-20%, and overlapping ranges therein) of the gel formulation.
  • the gelling agent may be present at about 0.5% to about 5% (w/w).
  • the gelling agent is present at about 1.25% (w/w).
  • the vehicle may be an aqueous solvent, such as purified water, which is used in an amount sufficient to bring the gel formulation to 100% (w/w) - e.g., purified water q.s.
  • a gel formulation for dermal delivery of SNA- 125 with reduced exposure includes about 5% w/w SNA-125, about 1% w/w benzyl alcohol, about 20% w/w/ propylene glycol, about 20% w/w DMSO, and about 1.25% w/w HEC, where trolamine is added q.s. to adjust the pH to between about 5.0 and about 7.5, and purified water q.s.
  • Certain embodiments of SNA-125 gel formulations provided herein are set forth in Table 1.
  • a method of treating a skin condition associated with TrkA, Janus Kinase (JAK1, JAK2 and/or JAK3), Tyrosine Kinase 2 (TYK2), Mitogen- Activated Protein Kinase (MAP2K and/or MAP3K) signaling is disclosed in accordance with another embodiment.
  • the method comprises: applying, or instructing application of, a topical gel formulation to a skin region, wherein the gel formulation fully or partially inhibits signaling in the skin region, and thereby treats the skin condition, wherein the gel formulation comprises: a polymer conjugate of an indolocarbazole compound, a preservative/solvent, a solvent, a penetration enhancer, a pH adjusting agent, a gelling agent, and a vehicle.
  • the polymer conjugate in the gel formulation is SNA- 125; in a variation, the particular SNA- 125 gel formulation is set forth above in Table 1.
  • Other reduced exposure compounds disclosed herein may be used with or instead of SNA- 125.
  • the skin condition is inflammation, pain, atopic dermatitis, psoriasis, pruritus associated with psoriasis, or another dermatologic condition.
  • Figure 1 depicts the dermal levels of CT340 (calculated ng/cm 2 ) in an ex vitro percutaneous absorption study with 5% w/w CT340 gel prototypes.
  • Figure 2 depicts the epidermal levels of CT340 (calculated ng/cm 2 ) in an ex vitro percutaneous absorption study with 5% w/w CT340 gel prototypes.
  • Figure 3 depicts combined dermal and epidermal levels of CT340 (calculated ng/cm 2 ) in an ex vitro percutaneous absorption study with 5% w/w CT340 gel prototypes.
  • Figure 4 shows representatives of two possible scenarios for the assignment of test fields as referred to in Example 4.
  • Figure 8 shows the relative percent improvement in disease transcriptome of day 12 lesional tissue versus day 1 lesional tissue as referred to in Example 4. Histogram groupings are, left to right, IMP1 (2 % SNA-125), IMP2 (0.2 % SNA-125), IMP3 (0.5 % SNA-120), and IMP4 (vehicle).
  • Effective delivery of pharmacologically active agents may be hindered by unwanted exposure of those agents to non-desired locations (such as the systemic circulation and/or lymphatic system).
  • topical agents useful in treating various skin disorders may result in toxic side effects because of systemic exposure.
  • One issue with delivering compositions comprising one or more active agents topically (or non-topically) is the concern that such agents need to be delivered in an amount and at a location sufficient to have a therapeutic effect.
  • exposure e.g., absorption or longevity of the composition in the systemic circulation, lymphatic system, or other non-targeted sites
  • compositions described herein are both therapeutically efficacious and minimize non-target (e.g., systemic or bloodstream) exposure.
  • the active agents are PEGylated or otherwise coupled to large molecules, and surprisingly, are effective in crossing biological membranes such that the active agents are effectively delivered to the target location. Inflammatory and non-inflammatory conditions are contemplated herein.
  • Reduced exposure compounds and compositions are provided in several embodiments.
  • “Reduced exposure” compounds are those compounds that, when delivered to a target location, are formulated to act at the target location with reduced exposure (e.g., entry and/or longevity) in non-target sites. Exposure is reduced as compared to active agents not formulated according to the embodiments described herein. As a non-limiting example, a PEGylated topical dermal active agent has reduced exposure to the bloodstream as compared to the active agent alone.
  • Reduced exposure compounds include topical compounds that can be delivered to body surfaces and cavities such as the skin, eyes, ears, nose, mouth, vagina, rectum, etc.
  • Non-desired target sites include, for example, the systemic system, the lymphatic system, non-target tissue, etc.
  • “Reduced exposure compositions” comprise or consist essentially of one or more “reduced exposure compounds.”
  • Reduced exposure topical compositions are provided in many embodiments.
  • less or none of the active agent is absorbed into the non target site (e.g., systemic circulation and/or lymphatic system).
  • the non target site e.g., systemic circulation and/or lymphatic system.
  • clearance e.g., by the kidney
  • One or more of the advantages of (i) reduced absorption into the non-target site (e.g., systemic circulation and/or lymphatic system), (ii) slower absorption into the non-target site (e.g., systemic circulation and/or lymphatic system), and (iii) faster clearance rates from the non-target site (e.g., systemic circulation and/or lymphatic system) are also achieved when using the compositions (e.g., via dermal topical formulations as described herein) for treating the skin.
  • a polymer conjugate comprising a warhead (e.g., at least one active agent) linked to a polymer, wherein the warhead comprises an indolocarbazole compound.
  • the polymer conjugate comprises an indolocarbazole compound of formula (I) or of formula (II):
  • R 1 and R 2 are the same or a different residue and are each independently selected from the group consisting of:
  • R 7 is selected from the group consisting of hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, - (CH 2 ) a C0 2 R 10 (wherein a is 1 or 2, and wherein R 10 is selected from the group consisting of hydrogen and substituted or unsubstituted lower alkyl) and -(CH 2 ) a C0 2 NR 5 R 6 ,
  • R 8 is selected from hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl
  • R 3 is hydrogen, halogen, acyl, carbamoyl, substituted or unsubstituted lower alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted lower alkynyl or amino;
  • W 1 and W 2 are independently hydrogen, hydroxy or W 1 and
  • X is a polymer moiety, either linear or branched
  • A represents - ⁇ C' and B represents -L 2 - Y', wherein at least one of X' and Y' is a polymer moiety, either linear or branched, which is bound by L 1 and/or L 2 to the tetrahydrofuran ring of the compound of formula (II); L 1 and/or L 2 are a covalent chemical bond or a linker group;
  • R 19 or R 20 are each independently selected from hydrogen, lower alkyl, lower alkenyl, lower alkynyl or R 19 or R 20 are independently the residue of an a-amino acid in which the hydroxy group of the carboxyl group is excluded, or R 19 or R 20 are combined with a nitrogen atom to form a heterocyclic group; and
  • the polymer moiety X, X' or/and Y' covalently attached to the indolocarbazole compound of formulae (I) and (II) has to be biocompatible, can be of natural or semi-synthetic or synthetic origin and can have a linear or branched structure.
  • the polymer moiety X, X' or/and Y' is selected from poly(alkylene oxides), in particular from (polyethylene) oxides.
  • polymers include without limitation polyacrylic acid, polyacrylates, polyacrylamide or N-alkyl derivatives thereof, polymethacrylic acid, polymethacrylates, polyethylacrylic acid, polyethylacrylates, polyvinylpyrrolidone, poly(vinylalcohol), polyglycolic acid, polylactic acid, poly(lactic-co-glycolic) acid, dextran, chitosan, polyaminoacids, hydroxyethyl starch.
  • the polymer moiety X, X' or/and Y' is a polyethylene glycol (PEG) moiety, wherein the terminal OH group can optionally be modified e.g. with C 1 -C 5 alkyl or C 1 -C 5 acyl groups.
  • the terminal OH group is optionally modified with Ci-, C 2 - or C 3 -alkyl groups or Ci-, C 2 - or C 3 groups.
  • the modified polyethylene glycol is a terminally alkoxy-substituted polyethylene glycol.
  • the polymer moiety is methoxy-polyethylene- glycol (mPEG).
  • the term“about” will be understood by one of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term“about” is meant to encompass variations of ⁇ 20 % or ⁇ 10 %, including ⁇ 5 %, ⁇ 1 %, and ⁇ 0.1 % from the specified value, as such variations are appropriate to perform the disclosed methods.
  • lower alkyl when used alone or in combination with other groups, means a straight chained or branched lower alkyl group containing from 1 -6 carbon atoms, preferably from 1-5, more preferably from 1-4 and especially preferably 1- 3 or 1-2 carbon atoms.
  • These groups include, in some embodiments, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, amyl, isoamyl, neopentyl, 1 -ethylpropyl, hexyl, and the like.
  • lower alkyl moiety of the "lower alkoxy”, the “lower alkoxycarbonyl”, the “lower akylaminocarbonyT, “lower hydroxyalkyl' and of the "tri-lower alkylsilyT groups has the same meaning as "lower alkyl” defined above.
  • the "lower alkenyl” groups are defined as CVG, alkenyl groups which may be straight chained or branched and may be in the Z or E form. Such groups include vinyl, propenyl, 1 -butenyl, isobutenyl, 2-butenyl, 1 -pentenyl, (Z)-2- pentenyl, (E)-2- pentenyl, (Z)-4-methyl-2-pentenyl, (E)-4-methyl-2-pentenyl, pentadienyl, e.g., 1 , 3 or 2,4-pentadienyl, and the like.
  • the C2-C6- alkenyl groups are C2-C5- , C2-C4-alkenyl groups. In other embodiments, the C2-C6- alkenyl groups are C2-C3- alkenyl groups.
  • lower alkynyl groups refers to C2-C6-alkynyl groups which may be straight chained or branched and include ethynyl, propynyl, 1 -butynyl, 2- butynyl, 1 -pentynyl, 2-pentynyl, 3-methyl-l -pentynyl, 3-pentynyl, 1 -hexynyl, 2- hexynyl, 3-hexynyl and the like.
  • C2-C6-alkynyl groups are C2-C5-, C2-C4-alkynyl groups.
  • C2-C6-alkynyl groups are C2-C3-alkynyl groups.
  • aryl group refers to C 6 -C 14-aryl groups which contain from 6 up to 14 ring carbon atoms. These groups may be mono-, bi- or tricyclic and are fused rings. In some embodiments, the aryl groups include phenyl, biphenyl, naphthyl, anthracenyl, phenanthrenyl and the like. The aryl moiety of the "arylcarbonyl” and the “arylaminocarbonyl” groups has the same meaning as defined above.
  • heteroaryl groups may contain 1 to 3 heteroatoms independently selected from nitrogen, sulfur or oxygen and refers C3-C 13-heteroaryl groups. These groups may be mono-, bi- or tricyclic.
  • the C3- C heteroaryl groups include heteroaromatics and saturated and partially saturated heterocyclic groups. These heterocyclics may be monocyclic, bicyclic, tricyclic.
  • the 5 or 6-membered heterocyclic groups are thienyl, furyl, pyrrolyl, pyridyl, pyranyl, morpholinyl, pyrazinyl, methyl pyrrolyl, and pyridazinyl.
  • the C3-C13- heteroaryl may be a bicyclic heterocyclic group.
  • the bicycbc heterocyclic groups are benzofuryl, benzothienyl, indolyl, imidazolyl, and pyrimidinyl.
  • the C3-Ci3-heteroaryls are furyl and pyridyl.
  • lower alkoxy includes alkoxy groups containing from 1 to 6 carbon atoms, in some embodiments from 1 to 5, in other embodiments from 1 -4 and in yet other embodiments 1 to 3 or 1 to 2 carbon atoms and may be straight chained or branched. These groups include methoxy, ethoxy, propoxy, butoxy, isopropoxy, tert- butoxy, pentoxy, hexoxy and the like.
  • acyl includes lower alkanoyl containing 1 to 6 carbon atoms, in some embodiments from 1 to 5, from 1 to 4, from 1 to 3 or from 1 to 2 carbon atoms and may be straight chained or branched. These groups include, in some embodiments, formyl, acetyl, propionyl, butyryl, isobutyryl, tertiary butyryl, pentanoyl and hexanoyl.
  • the acyl moiety of the "acyloxy" group has the same meaning as defined above.
  • halogen includes fluoro, chloro, bromo, iodio, and the like.
  • aralkyl refers CVCi 5-aralkyl wherein the alkyl group is substituted by an aryl.
  • the alkyl group and aryl may be selected from the Ci-C 6 alkyl groups and the C 6 -C 14-aryl groups as defined above, wherein the total number of carbon atoms is between 7 and 15.
  • the CVC 15-aralkyl groups are benzyl, phenylethyl, phenylpropyl, phenylisopropyl, phenylbutyl, diphenylmethyl, 1 , 1 - diphenylethyl, 1 ,2-diphenylethyl.
  • the aralkyl moiety of the "aralkyloxy" groups has the same meaning as defined above.
  • the substituted lower alkyl, alkenyl and alkynyl groups have 1 to 3 independently selected substituents, such as lower alkyl, hydroxy, lower alkoxy, carboxyl, lower alkoxycarbonyl, nitro, halogen, amino, mono- or di- lower alkylamino, dioxolane, dioxane, dithiolane, and dithione.
  • the lower alkyl substituent moiety of the substituted lower alkyl, alkenyl and alkynyl groups, and the lower alkyl moiety of the lower alkoxy, the lower alkoxycarbonyl, and the mono- or di-lower alkylamino substituents of the substituted lower alkyl, alkenyl and alkynyl groups have the same meaning as "lower alkyl” defined above.
  • the substituted aryl, the substituted heteroaryl and the substituted aralkyl groups each has 1 to 3 independently selected substituents, such as lower alkyl, hydroxy, lower alkoxy, carboxy, lower alkoxycarbonyl, nitro, amino, mono- or di-lower alkylamino, and halogen.
  • substituents such as lower alkyl, hydroxy, lower alkoxy, carboxy, lower alkoxycarbonyl, nitro, amino, mono- or di-lower alkylamino, and halogen.
  • the lower alkyl moiety of the lower alkyl, the lower alkoxy, the lower alkoxycarbonyl, and the mono- or di- lower alkylamino groups among the substituents has the same meaning as Tower alkyl’ defined above.
  • the heterocyclic group formed by R 5 and R 6 combined with a nitrogen atom includes pyrrolidinyl, piperidinyl, piperidino, morpholinyl, morpholino, thiomorpholino, N-methylpiperazinyl, indolyl, and isoindolyl.
  • each of R 1 and R 2 is hydrogen.
  • the residue R 14 is selected from phenyl, pyridyl, imidazolyl, thiazolyl, tetrazolyl, -COOR 15 , -OR 15 (wherein R 15 is in some embodiments selected from hydrogen, methyl, ethyl, phenyl or acyl), -SR 7 (wherein R 7 is in some embodiments selected from substituted or unsubstituted lower alkyl, 2-thiazoline and pyridyl) and -NR 5 R 6 (wherein R 5 and R 6 are in some embodiments selected from hydrogen, methyl, ethyl, phenyl, carbamoyl and lower alkylaminocarbonyl).
  • the residue R 16 is selected from hydrogen, methyl, ethyl, phenyl, imidazole, thiazole, tetrazole, -COOR 15 , -OR 15 and -NR 5 R 6 (wherein the residues R 15 , R 5 and R 6 have the meanings as described above).
  • the residue R 7 is selected from the group consisting of substituted or unsubstituted lower alkyl, substituted or unsubstituted phenyl, pyridyl, pyrimidinyl, thiazole and tetrazole.
  • k is 2, 3 or 4
  • j is 1 or 2
  • m and n are independently 0 or 1.
  • R 3 is hydrogen or acetyl. Furthermore, in some embodiments, each W 1 and W 2 is hydrogen.
  • X' when Y' is a polymer moiety and X' is not a polymer moiety, X' is selected from carboxy, hydroxymethyl or a lower alkoxycarbonyl. In some embodiments X' is selected from methoxy carbonyl.
  • the warhead of the polymer conjugate is a derivative of K252a, which has the formula:
  • the polymer conjugate is SNA- 125 (also referred to as CT340), wherein the composition has the formula:
  • SNA-125 also referred to as CT340
  • a non-target site such as the systemic circulation and/or lymphatic system
  • exposure at a non-target site is less than 90%, 75%, 50%, 25%, 15%, 10%, 5% or 2% (or less) of the polymer conjugate as compared to a similar active entity that has not been produced according to the embodiments described herein.
  • desirable rate of clearance from the non-target site (e.g., systemic circulation and/or lymphatic system) for the compositions described herein is increased by at least 10%, 25%, 50%, or 75% or more as compared to non-conjugated controls.
  • a PEGylated active agent described herein not only penetrates the desired membranes to reach a desired target, but has reduced non-target exposure by at least 20-80% or more as compared to the non- PEGylated active agent.
  • blood concentrations measured post administration of the compositions described herein are less than about 0.1 ng/ml, less than 1 ng/ml, or less than 10 ng/ml after, e.g., 15 minutes, 30 minutes, 1 hour, 6 hours or 12 hours.
  • reduced exposure at non-target sites contributes to enhanced efficacy.
  • Efficacy may be enhanced because lower concentrations/amounts/dosing schedules are required to achieve the same or similar therapeutic efficacy at the target site (because, for example, the active ingredient stays at the desired target site for a longer time).
  • concentrations/amounts/dosing schedules are reduced by 25%-75% or more.
  • More rapid clearance rates of the active agent once in the non-target site(s) are also beneficial because this may allow for a higher concentration or more doses to be delivered. This is especially beneficial for active agents in which a subject would benefit from a higher dose but cannot tolerate the higher dose due to toxicity at the non-target site (e.g., systemic toxicity). Faster clearance rates would permit the desired higher dose to be delivered according to the desired schedule. For example, a subject may be able to tolerate daily doses rather than weekly doses because of the reduced exposure.
  • the active agents of the compositions described herein are measured in non-target sites (e.g., the systemic circulation and/or lymphatic system) at less than amounts found when the active agent is delivered without conjugation (e.g., less than 0.5%, 1% or 2% after 6 or 12 hours, as compared with 3-15% (e.g., 3-6%) when the active agent is delivered without conjugation).
  • non-target sites e.g., the systemic circulation and/or lymphatic system
  • the active agents of the compositions described herein are measured in non-target sites (e.g., the systemic circulation and/or lymphatic system) at less than 0.5%, 1% or 2% after 3-24 hours, as compared to an amount 2-20 times greater when the active agent is delivered without conjugation.
  • non-target sites e.g., the systemic circulation and/or lymphatic system
  • clearance of the compositions occurs within minutes of exposure to the non-target site (e.g., systemic circulation and/or lymphatic system), as opposed to hours.
  • 50% clearance of the conjugated polymer compounds occurs in less than 5 minutes, 15 minutes, 30 minutes, 1 hour, 6 hours, and 12 hours of exposure to the systemic circulation and/or lymphatic system. Clearance times of the conjugated polymer compounds are reduced by more than 25%, 50%, 75% and 90%, as compared to the non- conjugated active agents or other formulations. These reduced clearance times are beneficial to reduce toxicity and undesired side effects.
  • an active agent may be increasingly toxic as it is metabolized in the non-target site (e.g., systemic circulation and/or lymphatic system) because the metabolites exhibit more toxicity than the original agent.
  • the non-target site e.g., systemic circulation and/or lymphatic system
  • faster clearance rates in some cases even before the toxic metabolites are created, are especially beneficial.
  • the term“active entity” or“active agent” as used herein should not be understood as limiting the participation of the polymer itself and/or the chemical linking moiety between the polymer and the warhead in defining the pharmacology of the polymer conjugate.
  • the polymer influences the selectivity and/or inhibitory activity of the polymer conjugate.
  • the chemical linking moiety between the polymer and warhead influences the selectivity and/or inhibitory activity of the polymer conjugate.
  • the polymer conjugates exhibit no change in selectivity or inhibitory activity against the therapeutic target in comparison with the unconjugated active agent.
  • the polymer conjugates exhibit a significant increase in selectivity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the polymer conjugates exhibit a significant increase in inhibitory activity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the polymer conjugates exhibit a significant increase in selectivity and inhibitory activity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the increased selectivity and/or inhibitory activity of the polymer conjugate against the therapeutic target in comparison with the unconjugated active agent causes decrease in undesired biological effects. In some embodiments, the increased selectivity of the polymer conjugate is caused by an increase of the hydrodynamic volume resulting from the conjugated polymer chain. In some embodiments, the polymer chain creates a higher steric hindrance which allows discrimination among the diverse shapes and sizes of the binding sites of different proteins, thus improving selectivity with respect to the active agent alone.
  • various inflammatory skin diseases are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1).
  • the inflammatory skin disease comprises, in some embodiments, psoriasis, psoriasis guttata, inverse psoriasis, pustular psoriasis, psoriatic erythroderma, pruritis associated with any of the various forms of psoriasis, acute febrile neutrophilic dermatosis, eczema, xerotic eczema, dyshidrotic eczema, vesicular palmar eczema, acne vulgaris, atopic dermatitis, contact dermatitis, allergic contact dermatitis, dermatomyositis, exfoliative dermatitis, hand eczema, pompholyx, keloids, rosacea, rosacea due to sarcoidosis, rosacea due
  • kits for treating psoriasis comprising administering an effective amount of a formulation provided in Table 1 to the subject.
  • provided herein are methods of treating atopic dermatitis in a subject in need thereof, comprising administering an effective amount of a formulation provided in Table 1 to the subject.
  • pruritis e.g., pruritis associated with psoriasis or atopic dermatitis
  • methods of treating pruritis comprising administering an effective amount of a formulation provided in Table 1 to the subject.
  • kits for treating psoriasis comprising administering an effective amount of a formulation comprising about 0.2 % to about 2.0 % w/w (e.g., about 0.2 %, about 0.5 %, about 1.0 %, or about 2.0 %) of SNA-125 to the subject.
  • kits for treating atopic dermatitis in a subject in need thereof comprising administering an effective amount of a formulation comprising about 0.2 % to about 2.0 % w/w (e.g., about 0.2 %, about 0.5 %, about 1.0 %, or about 2.0 %) of SNA-125 to the subject.
  • pruritis e.g., pruritis associated with psoriasis or atopic dermatitis
  • methods of treating pruritis comprising administering an effective amount of a formulation comprising about 0.2 % to about 2.0 % w/w (e.g., about 0.2 %, about 0.5 %, about 1.0 %, or about 2.0 %) of SNA- 125 to the subject.
  • the administration is once daily (e.g., once daily for 8 weeks or 12 weeks). In some embodiments, the administration is twice daily (e.g., twice daily for 8 weeks or 12 weeks).
  • the subject has a weekly mean I-NRS score of at least 5 on the 11 point I-NRS scale.
  • the subject has a weekly mean I-NRS score of at least 5 on the 11 point I-NRS scale and the psoriasis is mild-to-moderate psoriasis (e.g., mild-to-moderate plaque psoriasis).
  • the subject has mild-to-moderate psoriasis with at least moderate itch.
  • the psoriasis is mild to moderate psoriasis.
  • the psoriasis is mild to moderate plaque psoriasis.
  • various skin neoplasias are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1).
  • the skin neoplasia comprises, in some embodiments, squamous cell carcinoma, basal cell carcinoma, malignant melanoma, malignant cutaneous lymphoma, Kaposi's sarcoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • various vascular tumors are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1).
  • the vascular tumor comprises, in some embodiments, hemangiomas, Kaposi's sarcoma, lymphangioma, glomangioma, angiosarcoma, hemangioendothelioma, and infantile hemangiomas.
  • various bullous diseases are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1).
  • the bullous disease comprises, in some embodiments, bullous pemphigoid, erythema multiforme, dermatitis herpetiformis, epidermolysis bullosa acquisita, linear Immunoglobulin A disease, mucous membrane pemphigoid, pemphigoid gestationis, pemphigus foliaceus, and pemphigus vulgaris.
  • hair growth and cycling are modulated (e.g., modulated by administering an effective amount of a formulation provided in Table 1).
  • alopecia is treated.
  • the polymer conjugates are administered in combination with UV irradiation therapy.
  • polymer conjugates wherein the polymer is polyethylene glycol (PEG) or methoxy-polyethylene glycol (m- PEG).
  • PEG polyethylene glycol
  • m- PEG methoxy-polyethylene glycol
  • a pharmaceutical composition comprising or consisting essentially of a polymer conjugate disclosed herein that is formulated for topical administration.
  • methods of making and using the compositions described herein are provided.
  • the invention comprises a reduced exposure composition comprising at least one active entity linked to at least one polymer, wherein the composition has reduced exposure at a non-target site as compared to the active entity delivered without the polymer.
  • the non-target site comprises the systemic system, the lymphatic system and/or another non-target tissue site in some embodiments.
  • the active entity binds to a kinase. In some embodiments, the active entity binds to Tropomyosin receptor kinase A (TrkA). In some embodiments, the active entity binds to a Janus Kinase (JAK) family member in some embodiments. In some embodiments, the active entity binds to one or more of Janus Kinase 1 (JAK1), Janus Kinase 2 (JAK2), Janus Kinase 3 (JAK3), and/or Tyrosine kinase 2 (TYK2) in some embodiments.
  • JK1 Janus Kinase 1
  • JAK2 Janus Kinase 2
  • JAK3 Janus Kinase 3
  • TYK2 Tyrosine kinase 2
  • the active entity binds to mitogen-activated protein kinase kinase 2 (MAP2K) and/or mitogen-activated protein kinase kinase 3 (MAP2K3).
  • MAP2K mitogen-activated protein kinase kinase 2
  • MAP2K3 mitogen-activated protein kinase kinase 3
  • Such kinase binding by the active entity may be partially or fully inhibitory or not.
  • the polymer used in the reduced exposure compounds comprises polyethylene glycol (PEG) and/or methoxy-polyethylene glycol (m-PEG).
  • PEG polyethylene glycol
  • m-PEG methoxy-polyethylene glycol
  • the active entity has one or more carboxyl, hydroxyl, amino and/or sulfhydryl groups
  • the active entity is PEGylated (or conjugated/coupled to another polymer) at one or more of said carboxyl, hydroxyl, amino and/or sulfhydryl groups.
  • the reduced exposure compositions described herein are formulated for topical administration in several embodiments.
  • methods of treating one or more of the following are provided: inflammatory skin disease, vascular tumors, skin neoplasia, bullous diseases, alopecia, wounds, scars, autoimmune disorders, and cancerous or pre-cancerous lesions.
  • Methods for modulating hair growth and cycling are provided in some embodiments.
  • a method of treating any of the above-mentioned skin diseases or any other skin condition in need of treatment includes: applying, or instructing application of, a topical formulation (e.g., a formulation provided in Table 1) to a skin region, wherein the formulation fully or partially inhibits signaling in the skin region, and thereby treats the skin condition, wherein the formulation comprises: a polymer conjugate of an indolocarbazole compound in an oil-in-water emulsion.
  • the topical formulation applied in accordance with the method may be any of the gel formulations of SNA-125 as more fully described herein.
  • compositions provided herein may be administered via at least two routes of administration, either simultaneously or sequentially according to some embodiments.
  • the composition is administered via a first (e.g. topical dermal) route to a subject, wherein the subject further receives an additional agent via a second (e.g., non- dermal) route to achieve synergetic effects.
  • the active agent (SNA-125/CT340) may be synthesized as follows:
  • K252b 1 molar equivalent of K252b was pre-dried by suspending in 10 vol of dichloromethane and concentrating to dryness on the rotary evaporator at ⁇ 25°C then holding under reduced pressure to afford a yellowish solid.
  • the dried K252b was re suspended in 160 vol of dichloromethane at ambient temperature ( ⁇ 20°C) and 2 molar equivalents of 4-methylmorpholine were added. The resulting suspension was stirred for ⁇ 35-45 minutes.
  • the product from Step 2 is purified by preparative column chromatography using silica gel which has been pretreated with ethyl acetate containing 5% triethylamine.
  • the Step 2 product was dissolved in dichloromethane/ethyl acetate for application to the column. Elution with ethyl acetate enabled residual K252a and UV- inactive species to be eluted (as monitored by TLC).
  • the column was then conditioned with dichloromethane and eluted with dichloromethane/MeOH (98.5: 1.5 - 95.5:4.5). Fractions were collected and analyzed by HPLC and TLC, and pooled according to their purity. Evaporation of the combined desired product fractions led to CT340 of purity >95% by HPLC area.
  • a precipitation step is performed to transform it from a semisolid concentrate into a readily handled free-flowing solid.
  • the active agent is formulated for topical delivery.
  • the active agent may be a reduced exposure composition.
  • the active agent is a polymer conjugate of an indolocarbazole compound, thereby providing a reduced exposure indolocarbazole compound.
  • the polymer conjugate is the indolocabazole depicted in Formula (II).
  • the polymer conjugate is SNA-125 (formerly referred to as CT340; both terms are used interchangeably herein).
  • the topical delivery formulation can be in any form, including for example, an ointment, a gel, a balm, a cream, a lotion, a primer, a serum, a liquid, a spray, etc.
  • the topical delivery formulation is a gel.
  • the weight percentage (w/w) of active agent to the total weight of the topical delivery formulation may range from about 0.001% to about 20%, from about 0.005% to about 15%, from about 0.01% to about 12%, or from about 0.1% to about 10%, including any weight percentages within the disclosed ranges. In other embodiments, the weight percentage (w/w) of the active agent may be greater than or equal to about 1.00% w/w. In certain embodiments, the weight percentage (w/w) of the active agent to the total weight of the topical delivery formulation may fall within any range defined by any two of the above weight percentages.
  • the weight percentage (w/w) of the active agent may be less than or equal to about 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% w/w (e.g., less than or equal to about 5.0%, 2.0%, 1.0%, 0.5%, or 0.2%).
  • the weight percentage (w/w) of the active agent to the total weight of the topical delivery formulation may fall within any range defined by any two of the above weight percentages.
  • the active agent is a polymer conjugate of an indolocarbazole compound providing a reduced exposure.
  • the polymer conjugate is the indolocabazole depicted in Formula (II).
  • the polymer conjugate is SNA- 125.
  • a gel formulation includes: a polymer conjugate of an indolocarbazole compound, a preservative/solvent, a solvent, a penetration enhancer, a pH adjusting agent, a gelling agent, and a vehicle.
  • the polymer conjugate of an indolocarbazole compound is SNA-125.
  • the SNA-125 may be present at about 0.05% to about 25% (w/w) of the gel formulation. In a variation, the SNA-125 may be present at about 0.5% to about 10% (w/w) of the gel formulation. In another variation, the SNA-125 may be present at about 5% (w/w) of the gel formulation.
  • the preservative/solvent may be one or more selected from benzoic acid, sorbic acid, boric acid, methylparaben, ethylparaben, propylparaben, butylparaben, sodium benzoate, sodium propionate, potassium sorbate, chlorobutanol, benzyl alcohol and phenyl ethyl alcohol, phenol, chlorocresol, o-phenyl phenol, benzalkonium chloride, cetyl pyridinium chloride, imidurea, thimerisal, propyl gallate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), citric acid, ascorbic acid, ascorbyl palmitate, alpha-tocopherol and and disodium edetate.
  • benzoic acid sorbic acid
  • boric acid methylparaben
  • methylparaben ethylparaben
  • propylparaben propyl
  • the preservative/solvent is benzyl alcohol.
  • the preservative/solvent may be present at about 0.1% to about 10% (w/w) of the gel formulation, including any concentrations defined by the range.
  • the preservative/solvent may be present at about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, or 10% (w/w), and may fall within any range defined by any two of the above weight percentages.
  • the preservative/solvent may be present at about 1% (w/w) of the gel formulation.
  • the solvent may be one or more selected from ethanol, propylene glycol, glycerin and polyethylene glycol.
  • the solvent is propylene glycol.
  • the solvent may be present at about 1% to about 30% (w/w) of the gel formulation, including any concentrations defined by the range. In a variation, the solvent may be present at about 1%, 5%, 10%, 15%, 20%, 25% or 30% (w/w), and may fall within any range defined by any two of the above weight percentages. Alternatively, the solvent may be present at about 20% (w/w) of the gel formulation.
  • the penetration enhancer may be one or more selected from dimethylsulfoxide, Transcutol, oleic acid, azone, pyrrolidones, surfactants (e.g. SLS, Tween), urea, essential oils, terpenes and terpenoids, and phospholipids.
  • the penetration enhancer is dimethylsulfoxide (‘DMSO’).
  • DMSO dimethylsulfoxide
  • the penetration enhancer may be present at about 1% to about 30% (w/w) of the gel formulation, including any concentrations defined by the range.
  • the solvent may be present at about 1%, 5%, 10%, 15%, 20%, 25% or 30% (w/w), and may fall within any range defined by any two of the above weight percentages.
  • the penetration enhancer may be present at about 20% (w/w) of the gel formulation.
  • the pH adjusting agent may be one or more selected from sodium hydroxide, hydrochloric acid, phosphoric acid, and triethylamine.
  • the pH adjusting agent is trolamine.
  • the pH adjusting agent may be used if necessary in an amount sufficient to adjust the final pH of the gel formulation (‘q.s.’) to greater than or equal to about 5.0.
  • the final pH may be adjusted (‘q.s.’) to between about 5.0 and about 7.5, or to any pH selected from about 5, 5.2, 5.4, 5.6, 5.8, 6.0, 6.2, 6.4, 6.6, 6.8, 7.0, 7.2, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0.
  • the gelling agent may be one or more selected from hydroxyethyl cellulose, hydroxypropyl methylcellulose, carbopol, gelatin, sodium alginate, sodium carboxymethylcellulose, gelatin, polysaccharides, and silica.
  • the gelling agent is hydroxyethyl cellulose (‘HEC’) ⁇
  • HEC hydroxyethyl cellulose
  • the gelling agent may be present at about 0.05% to about 20% (w/w) of the gel formulation. Alternatively, the gelling agent may be present at about 0.5% to about 5% (w/w), including any concentrations defined by the range.
  • the gelling agent may be present at about 0.50%, 0.75%, 1.0%, 1.25%, 1.5%, 1.75%, 2.0%, 2.25%, 2.5%, 2.75%, 3.0%, 3.25%, 3.5%, 3.75%, 4.0%, 4.25%, 4.5%, 4.75% or 5.0% (w/w), and may fall within any range defined by any two of the above weight percentages.
  • the gelling agent may be present at about 1.25% (w/w) of the gel formulation.
  • the vehicle may be an aqueous solvent, such as purified water, which is used in an amount sufficient to bring the gel formulation to 100% (w/w) - i.e., purified water q.s.
  • the gel formulation is one of the gel formulations of Table 1.
  • a gel formulation for dermal delivery of SNA-125 with reduced exposure is disclosed, wherein the gel formulation includes about 5% w/w SNA-125, about 1% w/w benzyl alcohol, about 20% w/w/ propylene glycol, about 20% w/w DMSO, and about 1.25% w/w HEC, where trolamine is added q.s. to adjust the pH to between about 5.0 and about 7.5, and purified water q.s.
  • An embodiment of such an SNA- 125 gel formulation is set forth above in Table 1.
  • the purpose of this solubility study was to identify appropriate solvents and/or solvent blends suitable for the development of diverse dosage forms.
  • the target dosage forms were aqueous gels, lipophilic gels and petrolatum-based ointments. Only solvents present in the FDA's Inactive Ingredients Guide (IIG) for approved topical products were considered. Solubility was assessed by visual determination.
  • IIG Inactive Ingredients Guide
  • API (-0.25 g) was weighed into type 1 clear glass vials. To each vial, approximately 4.75 g of the solvent or solvent blend was added. Samples were shaken manually and visually observed to check if the API had dissolved. Clear solution indicated that the drug had dissolved. The resultant solutions were stored at 5°C and 40°C for two weeks. These samples were evaluated for their color, the clarity of the solution and assayed for CT340 content.
  • Results of the compatibility testing are provided in Table 2 below. Data from the experiments indicate that the drug can be dissolved at 5% w/w in a variety of solvent blends, permitting the development of diverse dosage forms. All solutions were light yellow in color initially. After 2 weeks at 5°C and 40°C, alcoholic solutions had remained unchanged, while the presence of Transcutol resulted in a darker yellow solution at 40°C. Other experimental solvent exhibited a slightly more intense yellow compared to the initial. The drug substance remained dissolved in all blends after two weeks at both 5°C and 40°C temperatures. CT340 appears to be compatible with all solvents tested other than Transcutol.
  • the drug substance CT340 does not exhibit significant solubility in non-polar solvents alone such as isopropyl myristate and caprylic capric triglyceride. However, it exhibits sufficient solubility in alcohol to make it possible to develop prototypes containing alcohol and non-polar solvents. Examples of these are seen in Table 2 (3650-6A and 6B). While the concentration of alcohol used in those blends is below the maximum listed in the IIG, it was desirable to lower it in the CT340 prototypes to minimize its potential sensory effect on the skin during the clinical trials.
  • the solvent blend for this prototype consisted mainly of purified water, DMSO and propylene glycol. Both hydroxyethylcellulose (HEC) and hydroxypropyl methylcellulose (HPMC) are commonly used to gel blends of water and propylene glycol. Both polymers are also in common use in topical products. However, the effectiveness of these polymers in gelling aqueous systems containing DMSO was unknown. It was therefore decided to test the gelling ability of HEC and HPMC using water and propylene glycol containing DMSO at the highest proposed concentration (40% w/w). The test compositions are provided in Table 4.
  • Composition 10A was a clear viscous gel while 10B was a 'lumpy' runny product.
  • HEC was selected for use as the gelling agent.
  • the solvent composition for this prototype consisted of Isopropyl Myristate, Oleic Acid, Caprylic Capric Triglyceride, Alcohol, Propylene Glycol and Benzyl Alcohol.
  • Quantitative compositions of the five prototypes are provided in Table 5.
  • the three aqueous gels are all thickened with hydroxy ethylcellulose.
  • the three differ from each other primarily in their concentrations of DMSO, propylene glycol and benzyl alcohol.
  • the lipophilic solution consists of a blend of polar and non-polar solvents.
  • the fifth prototype is a petrolatum based ointment in which the drug phase is dispersed in the waxy base.
  • the drug phase utilizes benzyl alcohol and propylene glycol to dissolve the drug substance.
  • This stability study is designed to determine the chemical and physical stability of CT340 5.0% w/w prototype formulations. These formulation prototypes were prepared as ⁇ 60g (gels) & lOOg (ointment) laboratory batches. All batches there then split to have approximately 5g in scintillation vials for stability study. The prototypes were stored at 25°C/60% RH, and 40°C/75%RH for up to 6 months. The storage conditions and testing schedule are depicted in Tables 6 and 7 below. For the packaging, glass vials were wrapped with aluminum foil. There were nine total vials per formulation. The sample orientation was vertical, and sample storage after pull and prior to testing was controlled room temperature. The following testing was performed: appearance (visual, single determination) and assay (duplicate determination).
  • J 25C samples were evaluated at 3 months; 40C samples were evaluated at 2 months.
  • Drug product efficacy is determined by controlled clinical trials. Bioavailability and potency of topically applied active pharmaceutical ingredients are key components in efficacious drug products. Potential cutaneous (local) and/or systemic bioavailability can be assessed using in vitro percutaneous absorption testing. Data generated using in vitro skin permeation models can support formulation selection during pharmaceutical development programs.
  • Topical formulations containing CT340 are under development for the treatment of pain.
  • the purpose of this study was to characterize in vitro percutaneous absorption of CT340 from topical formulations following application to excised human skin from elective surgery.
  • CT340GP3 G is the most efficient at delivering CT340 into the tissue layers while CT3400P5 (I) is the least efficient.
  • In vitro skin permeation data is intended to facilitate identification of formulation candidates with the highest potential of success, in terms of developing a topical formulation with appropriate delivery characteristics. This approach may also provide the highest potential for correlating in vitro permeation data with clinical efficacy in diseases where the site of action is located in the viable tissues of the skin.
  • the clinically relevant dose of 5 mg/cm 2 of formulation was applied to dermatomed human abdominal tissue from a single donor obtained following elective surgery.
  • the thickness of the tissue ranged from 0.022 - 0.033 inches (0.559 - 0.838 mm) with a mean +/- standard deviation in thickness of 0.028 +/- 0.003 inches (0.708 +/- 0.084 mm) and a coefficient of variation of 11.9%.
  • Percutaneous absorption was evaluated using this human abdominal tissue from a single donor mounted in Bronaugh flow-through diffusion cells.
  • the cells were maintained at a constant temperature of 32 °C by use of recirculating water baths. These cells have a nominal diffusion area of 0.64 cm 2 .
  • the formulation residing on the tissue surface was removed by tape-stripping with CuDerm D-Squame stripping discs. These have been retained for potential future analysis.
  • the epidermis, dermis, and receptor phase samples were labeled and frozen prior to subsequent analysis of CT340 content by LC/Fluorescence.
  • the tissue sample limit of quantification (LOQ) achieved by the R&D LC/Fluorescence analytical method for CT340 was 1.058 ng/mL.
  • LC/Fluorescence was selected as the detection technique for this study.
  • LC/MS was investigated, but was less sensitive than the LC/fluorescence technique.
  • Em 400 nm
  • Tissue permeation and deposition results were statistically evaluated using unpaired student's t-tests (significant differences between formulations were defined by a p-value of ⁇ 0.05, at the 95% confidence interval). Significant differences between formulations were defined by a p value of ⁇ 0.05.
  • Outlier tests were performed using Grubbs Test. Outliers in the tissue data whether detected in the epidermis data or in the dermis data were removed from the analysis of both tissue types.
  • the efficiency of delivery is characterized by expressing amount of drug permeating as a percent of applied dose.
  • CT340AhGP4 (H) and CT340GP3 (G) had the highest efficiency of CT340 dermal deposition with 6.59 and 8.41 percent of the applied dose, respectively.
  • formulation CT3400P5 (I) exhibited the least efficient delivery into the dermis.
  • CT340 dermal deposition [000174] The calculated CT340 dermal deposition ranged from 6,867 to 21,034 ng/cm 2 .
  • Formulations CT340AhGP4 (H) and CT340GP3 (G) generated the highest CT340 dermal deposition with 16,472 and 21,034 ng/cm 2 , respectively.
  • Formulation CT3400P5 (I) produced the lowest CT340 dermal deposition, 6,867 ng/cm 2 .
  • CT340 epidermal deposition ranged from 25,341 to 43,024 ng/cm 2 .
  • Formulations CT340GP2 (F) and CT340GP3 (G) had the highest CT340 epidermal deposition with 41,119 and 43,024 ng/cm 2 , respectively.
  • Formulation CT3400P5 (I) generated the lowest CT340 epidermal deposition, 25,341 ng/cm 2 .
  • Figure 3 presents the summed dermal and epidermal levels for all formulations used in this study.
  • In vitro skin permeation data is intended to facilitate identification of formulation candidates with the highest potential of success, in terms of developing a topical formulation with appropriate delivery characteristics. This approach may also provide the highest potential for correlating in vitro permeation data with clinical efficacy, in diseases where the site of action is located in the viable tissues of the skin.
  • EXAMPLE 3 EVALUATION OF THE SAFETY AND EFFICACY OF TOPICALLY APPLIED SNA-125 GEL IN HEALTHY VOLUNTEERS AND
  • the study subjects consisted of 2 sequential cohorts of adult male or female subjects: (1) healthy volunteers (HV) and (2) subjects with atopic dermatitis (AD).
  • HV healthy volunteers
  • AD subjects with atopic dermatitis
  • 4 HV were evaluated for the safety of topically applied SNA-125 gel and vehicle following 7 days of treatment.
  • the gel formulations (see Table 1) contain SNA- 125 (active ingredient), benzyl alcohol, propylene glycol, dimethylsulfoxide (DMSO), hydroxyethylcellulose (HEC), trolamine, and water (as needed) (the vehicle formulation was the same as the SNA-125 formulation without the active).
  • EXAMPLE 4 PHASE 1 RANDOMIZED. VEHICLE- AND COMPARATOR- CONTROLLED. DOUBLE-BLIND TRIAL TO ASSESS EFFICACY AND SAFETY OF SNA-125 IN A PSORIASIS PLAQUE TEST
  • Psoriasis is a common and often chronic skin disease that affects approximately 2 % of the population.
  • Chronic plaque psoriasis is the most common form, affecting 85 to 95 % of all patients with the disease. The rate of psoriasis varies according to age, gender, region and ethnicity.
  • Chronic plaque psoriasis is characterized by raised, well-demarcated, erythematous oval plaques covered with adherent silvery scales and is most commonly found on the scalp, nails, lower back and the extensor sides of the joints. The scales result from a hyperproliferative epidermis with premature maturation of keratinocytes and incomplete comification, with retention of nuclei in the stratum comeum.
  • the inflammatory infiltrate consists of dendritic cells, macrophages and T-cells in the dermis, with neutrophils and some T-cells in the epidermis. Abnormalities of the innate immune system are also involved.
  • SNA-125 represents one approach to the treatment (e.g., topical) of atopic dermatitis, psoriasis vulgaris, or both, as well as the substantial and undertreated condition of associated pruritus.
  • Two positive controls will also be tested in this study: SNA-120; and DAIVONEX® Ointment 50 pg (calcipotriol)— a topical treatment for psoriasis vulgaris.
  • SNA- 125 has been formulated for topical administration as an aqueous gel containing dimethyl sulfoxide (DMSO), propylene glycol, hydroxyethyl cellulose, benzyl alcohol, and trolamine at 2% w/w and 0.2% w/w strengths.
  • DMSO dimethyl sulfoxide
  • a matching placebo vehicle with the same quantitative/qualitative composition with the exclusion of the active component has also been developed.
  • the placebo vehicle was found to possess mild irritation potential in skin of subjects with a history of atopic dermatitis in a l4-day cumulative irritation assay.
  • One objective of this trial is to evaluate antipsoriatic efficacy and safety of SNA- 125 in topical formulations compared to topically applied reference formulations and vehicle in subjects with psoriasis vulgaris.
  • test fields located on the torso or the extremities will be examined per subject.
  • the test fields will be treated occlusively for a 12-day treatment period (no IMP application on Day 7 and 12).
  • Treatment will be topical application of approximately 100 pL per test field (1.1 cm 2 ) of the respective IMP once daily during a l2-day treatment period (10 treatments, no application on Days 7 and 12).
  • Investigational medicinal products to be administered are as follows: IMP 1 : 2% w/w SNA-125, daily dosage: approx. 2 mg SNA-125, total dosage for the treatment period: approx. 20 mg SNA- 125;
  • IMP 2 0.2% w/w SNA-125, daily dosage: approx. 0.2 mg SNA-125, total dosage for the treatment period: approx. 2 mg SNA-125;
  • IMP 1 and IMP 2 total combined dosage of SNA-125: daily dosage: approx. 2.2 mg SNA-125, total dosage for the treatment period: approx. 22 mg SNA-125; IMP 3: 0.5% w/w SNA-120 (comparator), daily dosage: approx. 0.5 mg SNA-120, total dosage for the treatment period: approx. 5 mg SNA- 120;
  • IMP 4 (vehicle): active ingredient-free vehicle to IMP 1 and IMP 2;
  • IMP 5 (marketed comparator): DAIVONEX® 50 pg/g ointment (calcipotriol 0.005%), daily dosage: approx. 0.005 mg calcipotriol, total dosage for the treatment period: approx. 0.05 mg calcipotriol.
  • test fields will be numbered with 1 to 5.
  • Figure 4 shows representatives of 2 of a number of possible scenarios. The actual distribution of plaques and test fields may vary individually. Randomization of the IMPs 1 to 5 will be performed using a Williams design. For each subject, a treatment sequence will be randomly assigned.
  • each IMP will be applied to a total of 5 test fields using special test chambers (Finn chambers, 12 mm inside 0, 14 mm outside 0). This is the amount required to completely fill the test chamber.
  • the IMPs will be applied in the Finn chamber and seated in holes punched in a hydrocolloid dressing.
  • the hydrocolloid dressing will be fixed before on the skin with adhesive patches containing the same holes for the chambers like the hydrocolloid dressing.
  • the chambers themselves will be fixed in place with LEUKOSILK® or the like and will be removed before each new application.
  • the distance between the chambers must be at least 1.5 cm to exclude interactions with neighboring fields.
  • the fields will be treated occlusively for a treatment period of 12 days. On 10 of the trial days treatments will be performed (Days 1 to 6 and 8 to 11). Before each new application, remaining preparation residues will be removed by gently cleansing each test field with a separate soft tissue.
  • the hydrocolloid dressing will be renewed on Days 4 and 8.
  • Blood pressure and pulse rate will be measured at screening, baseline and Day 12 (EoT). A physical examination will be performed at screening, Day 1 and on Day 12.
  • the trial is aimed to detect a treatment effect as compared to DAIVONEX® 50 pg/g ointment (calcipotriol 0.005%) within the planned trial design.
  • a former in-house trial revealed a psoriatic infiltrate thickness reduction on Day 12 of about 51 % of the baseline level with a coefficient of variance (CV) below 0.97 for the difference to vehicle in change from baseline.
  • CV coefficient of variance
  • biopsies were taken per subject. Four biopsies were taken at baseline from lesional and non-lesional skin. Eight biopsies were taken at Day 12 (EoT) from treated lesional skin including 2 from each test field for 2% and 0.2% SNA-125, gel vehicle, and 0.5% SNA-120.
  • H&E and immunohistochemistry (IHC) results from one subject are shown in panel display in Figure 5.
  • these data demonstrate an improvement on epidermal thickness and staining for various biomarkers. Specifically, 0.2 % SNA-125 had a reduction in epidermal thickness and K16 expression. Also, 0.5 % SNA-120 showed a reduction in K16 staining.
  • Figure 6 shows the aggregate epidermal thickness results for treated lesional skin at Day 12. There was a statistically significant improvement in epidermal thickness for 0.2 % SNA-125 compared to Baseline (day 1) lesional skin.
  • Figure 8 shows the results of disease (i.e. psoriasis) transcriptome (i.e., discrete set of co-modulated genes) analysis, quantification of improvement of different disease pathways. The best improvements were seen with 0.2 % SNA-125 and 0.5 % SNA- 120.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

Topical formulations to treat skin conditions, such as atopic dermatitis, psoriasis, pruritus, are provided in which a polymer conjugate of an indolocarbazole exhibits reduced systemic exposure. An example formulation includes the TrkA/JAK/MAP kinase antagonist, SNA-125, in a gel formulation.

Description

FORMULATIONS FOR DERMAL DELIVERY OF POLYMER CONJUGATES OF INDOLOCARBAZOLE COMPOUNDS WITH REDUCED EXPOSURE
RELATED APPLICATIONS
[0001] This application claims priority to U.S. Provisional Patent Application No. 62/608,366, filed December 20, 2017, U.S. Provisional Patent Application No. 62/723,347, filed August 27, 2018, U.S. Provisional Patent Application No. 62/732,538, filed September 17, 2018, and U.S. Provisional Patent Application No. 62/774,729, filed December 3, 2018. The entire content of each of these applications is incorporated herein by reference.
BACKGROUND
[0002] Skin care formulations, which include for example, primers, serums, lotions, ointments, gels, creams, foams and other products, are used on the skin for various purposes. Formulations can contain one or more active agents, including in some embodiments, indolocarbazole compounds conjugated to polymer(s).
SUMMARY
[0003] Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds, having reduced exposure, are disclosed. In one example, a gel formulation is disclosed. The gel formulation includes or consists essentially of, for example: a polymer conjugate of an indolocarbazole compound, a preservative/solvent, a solvent, a penetration enhancer, a pH adjusting agent, a gelling agent, and a vehicle.
[0004] In one embodiment of the gel formulation, the polymer conjugate of an indolocarbazole compound is SNA- 125 (formerly referred to as CT340; both terms are used interchangeably herein). The SNA-125 may be present at about 0.05% to about 25% (w/w) of the gel formulation. In a variation, the SNA-125 may be present at about 0.5% to about 10% (w/w) of the gel formulation (e.g., about 0.2-2%, 0.5-2%, 2-6%, or 6- 10%, e.g., about 0.2%, 1.0%, 2.0%, or 5.0%, and overlapping ranges therein). In another variation, the SNA-125 may be present at about 5% (w/w) of the gel formulation. Other reduced exposure compounds disclosed herein may be used with or instead of SNA-125.
[0005] In certain embodiments of the disclosed gel formulation, the preservative/solvent may be one or more selected from benzoic acid, sorbic acid, boric acid, methylparaben, ethylparaben, propylparaben, butylparaben, sodium benzoate, sodium propionate, potassium sorbate, chlorobutanol, benzyl alcohol and phenyl ethyl alcohol, phenol, chlorocresol, o-phenyl phenol, benzalkonium chloride, cetyl pyridinium chloride, imidurea, thimerisal, propyl gallate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), citric acid, ascorbic acid, ascorbyl palmitate, alpha-tocopherol and disodium edetate. In one embodiment, the preservative/solvent is benzyl alcohol. The preservative/solvent may be present at about 0.1% to about 10% (w/w) (e.g., 0.1-3%, 3-6%, 6-10%, and overlapping ranges therein) of the gel formulation. Alternatively, the preservative/solvent may be present at about 1% (w/w) of the gel formulation.
[0006] In certain embodiments of the disclosed gel formulation, the solvent may be one or more selected from water, ethanol, propylene glycol, glycerin and polyethylene glycol. In one embodiment, the solvent is propylene glycol. The solvent may be present at about 1% to about 30% (w/w) (e.g., 1-10%, 10-25%, 25-30%, and overlapping ranges therein) of the gel formulation. Alternatively, the solvent may be present at about 20% (w/w) of the gel formulation.
[0007] In certain embodiments of the disclosed gel formulation, the penetration enhancer may be one or more selected from dimethylsulfoxide (‘DMSO’), Transcutol, oleic acid, azone, pyrrolidones, surfactants (e.g. SLS, Tween), urea, essential oils, terpenes and terpenoids, and phospholipids. In one embodiment, the penetration enhancer is DMSO. The penetration enhancer may be present at about 1% to about 30% (w/w) (e.g., 1-10%, 10-25%, 25-30%, and overlapping ranges therein) of the gel formulation. Alternatively, the penetration enhancer may be present at about 20% (w/w) of the gel formulation.
[0008] In certain embodiments of the disclosed gel formulation, the pH adjusting agent may be one or more selected from trolamine, sodium hydroxide, hydrochloric acid, phosphoric acid, and triethylamine. In one embodiment, the pH adjusting agent is trolamine. The pH adjusting agent may be used in an amount sufficient to adjust the final pH of the gel formulation (‘q.s.’) to greater than or equal to about 5.0.
[0009] In certain embodiments of the disclosed gel formulation, the gelling agent may be one or more selected from hydroxyethyl cellulose, hydroxypropyl methylcellulose, carbopol, gelatin, sodium alginate, sodium carboxymethylcellulose, gelatin, polysaccharides and silica. In one embodiment, the gelling agent is hydroxyethyl cellulose (‘HEC’). The gelling agent may be present at about 0.05% to about 20% (w/w) (e.g., 0.05-5%, 6-15%, 15-20%, and overlapping ranges therein) of the gel formulation. Alternatively, the gelling agent may be present at about 0.5% to about 5% (w/w). In one embodiment of the gel formulation, the gelling agent is present at about 1.25% (w/w).
[00010] In certain embodiments of the disclosed gel formulation, the vehicle may be an aqueous solvent, such as purified water, which is used in an amount sufficient to bring the gel formulation to 100% (w/w) - e.g., purified water q.s.
[00011] In one embodiment, a gel formulation for dermal delivery of SNA- 125 with reduced exposure is disclosed, wherein the gel formulation includes about 5% w/w SNA-125, about 1% w/w benzyl alcohol, about 20% w/w/ propylene glycol, about 20% w/w DMSO, and about 1.25% w/w HEC, where trolamine is added q.s. to adjust the pH to between about 5.0 and about 7.5, and purified water q.s. Certain embodiments of SNA-125 gel formulations provided herein are set forth in Table 1.
Table 1.
Figure imgf000004_0001
[00012] A method of treating a skin condition associated with TrkA, Janus Kinase (JAK1, JAK2 and/or JAK3), Tyrosine Kinase 2 (TYK2), Mitogen- Activated Protein Kinase (MAP2K and/or MAP3K) signaling is disclosed in accordance with another embodiment. The method comprises: applying, or instructing application of, a topical gel formulation to a skin region, wherein the gel formulation fully or partially inhibits signaling in the skin region, and thereby treats the skin condition, wherein the gel formulation comprises: a polymer conjugate of an indolocarbazole compound, a preservative/solvent, a solvent, a penetration enhancer, a pH adjusting agent, a gelling agent, and a vehicle. In one embodiment of the disclosed method, the polymer conjugate in the gel formulation is SNA- 125; in a variation, the particular SNA- 125 gel formulation is set forth above in Table 1. Other reduced exposure compounds disclosed herein may be used with or instead of SNA- 125.
[00013] In one embodiment of the disclosed method, the skin condition is inflammation, pain, atopic dermatitis, psoriasis, pruritus associated with psoriasis, or another dermatologic condition.
BRIEF DESCRIPTION OF THE DRAWINGS
[00014] Figure 1 depicts the dermal levels of CT340 (calculated ng/cm2) in an ex vitro percutaneous absorption study with 5% w/w CT340 gel prototypes.
[00015] Figure 2 depicts the epidermal levels of CT340 (calculated ng/cm2) in an ex vitro percutaneous absorption study with 5% w/w CT340 gel prototypes.
[00016] Figure 3 depicts combined dermal and epidermal levels of CT340 (calculated ng/cm2) in an ex vitro percutaneous absorption study with 5% w/w CT340 gel prototypes.
[00017] Figure 4 shows representatives of two possible scenarios for the assignment of test fields as referred to in Example 4.
[00018] Figure 5 shows H&E and immunohistochemistry (IHC) results of Example 4 from a single subject in panel display; Dl NLS = day 1 non-lesional skin, Dl LS = day 1 lesional skin.
[00019] Figure 6 shows the aggregate epidermal thickness (mm) results for treated lesional skin at Day 12 in Example 4; Dl NL = day 1 non-lesional skin, Dl LS = day 1 lesional skin.
[00020] Figure 7 shows post-treatment lesional skin compared to baseline lesional skin of psoriasis transcriptome as referred to in Example 4 (left = down- regulated, right = up-regulated).
[00021] Figure 8 shows the the relative percent improvement in disease transcriptome of day 12 lesional tissue versus day 1 lesional tissue as referred to in Example 4. Histogram groupings are, left to right, IMP1 (2 % SNA-125), IMP2 (0.2 % SNA-125), IMP3 (0.5 % SNA-120), and IMP4 (vehicle). [00022] Figure 9 shows post-treatment lesional skin compared to baseline lesional skin of psoriasis transcriptome in responders as referred to in Example 4 (left = down-regulated, right = up-regulated).
DETAILED DESCRIPTION
[00023] Effective delivery of pharmacologically active agents may be hindered by unwanted exposure of those agents to non-desired locations (such as the systemic circulation and/or lymphatic system). For example, topical agents useful in treating various skin disorders may result in toxic side effects because of systemic exposure. One issue with delivering compositions comprising one or more active agents topically (or non-topically) is the concern that such agents need to be delivered in an amount and at a location sufficient to have a therapeutic effect. At the same time however, exposure (e.g., absorption or longevity of the composition in the systemic circulation, lymphatic system, or other non-targeted sites) may not be desirable for multiple reasons, including, but not limited to, safety reasons. There remains an unmet need for compounds with reduced exposure at non-target sites that result in a clinically therapeutic effect.
[00024] In several embodiments of the invention, the compositions described herein are both therapeutically efficacious and minimize non-target (e.g., systemic or bloodstream) exposure. In some embodiments, the active agents are PEGylated or otherwise coupled to large molecules, and surprisingly, are effective in crossing biological membranes such that the active agents are effectively delivered to the target location. Inflammatory and non-inflammatory conditions are contemplated herein.
[00025] Reduced exposure compounds and compositions are provided in several embodiments. “Reduced exposure” compounds are those compounds that, when delivered to a target location, are formulated to act at the target location with reduced exposure (e.g., entry and/or longevity) in non-target sites. Exposure is reduced as compared to active agents not formulated according to the embodiments described herein. As a non-limiting example, a PEGylated topical dermal active agent has reduced exposure to the bloodstream as compared to the active agent alone. Reduced exposure compounds include topical compounds that can be delivered to body surfaces and cavities such as the skin, eyes, ears, nose, mouth, vagina, rectum, etc. Non-desired target sites include, for example, the systemic system, the lymphatic system, non-target tissue, etc. “Reduced exposure compositions” comprise or consist essentially of one or more “reduced exposure compounds.”
[00026] Reduced exposure topical compositions are provided in many embodiments. In some cases, less or none of the active agent is absorbed into the non target site (e.g., systemic circulation and/or lymphatic system). Further, once the composition enters the systemic circulation and/or lymphatic system, clearance (e.g., by the kidney) occurs at a much faster rate. One or more of the advantages of (i) reduced absorption into the non-target site (e.g., systemic circulation and/or lymphatic system), (ii) slower absorption into the non-target site (e.g., systemic circulation and/or lymphatic system), and (iii) faster clearance rates from the non-target site (e.g., systemic circulation and/or lymphatic system) are also achieved when using the compositions (e.g., via dermal topical formulations as described herein) for treating the skin.
[00027] In several embodiments, there is provided in a reduced exposure composition, a polymer conjugate comprising a warhead (e.g., at least one active agent) linked to a polymer, wherein the warhead comprises an indolocarbazole compound. In some embodiments, the polymer conjugate comprises an indolocarbazole compound of formula (I) or of formula (II):
Figure imgf000007_0001
formula (I) formula (II)
[00028] wherein in formula (I) and (II)
[00029] R1 and R2 are the same or a different residue and are each independently selected from the group consisting of:
[00030] (a) hydrogen, halogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, hydroxy, lower alkoxy, carboxy, lower alcoxycarbonyl, acyl, nitro, carbamoyl, lower alkylaminocarbonyl, -NR5R6, wherein R5 and R6 are each independently selected from hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, substituted or unsubstituted lower alkylaminocarbonyl, substituted or unsubstituted lower arylaminocarbonyl, alkoxycarbonyl, carbamoyl, acyl or R5 and R6 are combined with a nitrogen atom to form a heterocyclic group,
[00031] (b) -CO(CH2)jR4, wherein j is 1 to 6, and R4 is selected from the group consisting of
[00032] (i) hydrogen, halogen, -N3,
[00033] (ii) -NR5R6, wherein R5 and R6 are as defined above,
[00034] (iii) -SR7, wherein R7 is selected from the group consisting of hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, - (CH2)aC02R10 (wherein a is 1 or 2, and wherein R10 is selected from the group consisting of hydrogen and substituted or unsubstituted lower alkyl) and -(CH2)aC02NR5R6,
[00035] (iv) -OR8, -OCOR8, wherein R8 is selected from hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl
[00036] (c) -CH(OH)(CH2)j R4 wherein j and R4 are as defined above;
[00037] (d) -(CH2)dCHRnC02R12 or -(CH2)dCHRnCONR5R6, wherein d is 0 to 5, R11 is hydrogen, -CONR5R6, or -C02R13, wherein R13 is hydrogen or a wherein substituted or unsubstituted lower alkyl, and R12 is hydrogen or a substituted or unsubstituted lower alkyl;
[00038] (e) -(CH2)kR14 wherein k is 2 to 6 and R14 is halogen, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, -COOR15, -OR15, (wherein R15 is hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or acyl), -SR7 (wherein R7 is as defined above), -CONR5R6, -NR5R6 (wherein R5 and R6are as defined above) or -N3; [00039] (f) -CH=CH(CH2)mR16, wherein m is 0 to 4, and R16 is hydrogen, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower alkenyl, substituted or unsubstituted lower alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, -COOR15, -OR15 (wherein R15 is as defined above) - CONR5R6 or -NR5R6 (wherein R5 and R6 are as defined above);
[00040] (g) -CH=C(C02R12)2, wherein R12 is as defined above;
[00041] (h) -CºC(CH2)nR16, wherein n is 0 to 4 and R16 is as defined above;
[00042] (i) -CH2OR22, wherein R22 is tri-lower alkyl silyl in which the three lower alkyl groups are the same or different or wherein R22 has the same meaning as R8
[00043] (j) -CH(SR23)2 and -CH2-SR7 wherein R23 is lower alkyl, lower alkenyl or lower alkynyl and wherein R7 is as defined above; and
[00044] R3 is hydrogen, halogen, acyl, carbamoyl, substituted or unsubstituted lower alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted lower alkynyl or amino; and
[00045] W1 and W2 are independently hydrogen, hydroxy or W1 and
W2 together represent oxygen;
[00046] and wherein in formula (I), X is a polymer moiety, either linear or branched,
[00047] and wherein in formula (II), A represents -ΐ C' and B represents -L2- Y', wherein at least one of X' and Y' is a polymer moiety, either linear or branched, which is bound by L1 and/or L2 to the tetrahydrofuran ring of the compound of formula (II); L1 and/or L2 are a covalent chemical bond or a linker group;
[00048] when Y' is a polymer moiety, and X' is not a polymer, L1 is a covalent chemical bond and X' is selected from the group consisting of
[00049] (a) hydrogen, lower hydroxyalkyl, acyl, carboxy, lower alkoxy carbonyl,
[00050] (b) -CONR17aR17b, wherein R17a and R17b are each independently selected from
[00051] (i) hydrogen, lower alkyl, lower alkenyl, lower alkynyl,
[00052] (ii) -CH2 R18; wherein R18 is hydroxy,
[00053] or (iii) -NR19R20, wherein R19 or R20 are each independently selected from hydrogen, lower alkyl, lower alkenyl, lower alkynyl or R19 or R20 are independently the residue of an a-amino acid in which the hydroxy group of the carboxyl group is excluded, or R19 or R20 are combined with a nitrogen atom to form a heterocyclic group; and
[00054] (c) -CH=N-R21, wherein R21 is hydroxy, lower alkoxy, amino, guanidino, or imidazolylamino;
[00055] when X' is a polymer moiety, and Y' is not a polymer, L2 is a covalent chemical bond and Y' is selected from hydroxy, lower alkoxy, aralkyloxy, or acyloxy;
[00056] or a pharmaceutically acceptable salt of formula (I) and/or (II).
[00057] The polymer moiety X, X' or/and Y' covalently attached to the indolocarbazole compound of formulae (I) and (II) has to be biocompatible, can be of natural or semi-synthetic or synthetic origin and can have a linear or branched structure. In some embodiments, the polymer moiety X, X' or/and Y' is selected from poly(alkylene oxides), in particular from (polyethylene) oxides. However, further exemplary polymers include without limitation polyacrylic acid, polyacrylates, polyacrylamide or N-alkyl derivatives thereof, polymethacrylic acid, polymethacrylates, polyethylacrylic acid, polyethylacrylates, polyvinylpyrrolidone, poly(vinylalcohol), polyglycolic acid, polylactic acid, poly(lactic-co-glycolic) acid, dextran, chitosan, polyaminoacids, hydroxyethyl starch.
[00058] In some embodiments, the polymer moiety X, X' or/and Y' is a polyethylene glycol (PEG) moiety, wherein the terminal OH group can optionally be modified e.g. with C1-C5 alkyl or C1-C5 acyl groups. In some embodiments, the terminal OH group is optionally modified with Ci-, C2- or C3-alkyl groups or Ci-, C2- or C3 groups. In some embodiments, the modified polyethylene glycol is a terminally alkoxy-substituted polyethylene glycol. In some embodiments, the polymer moiety is methoxy-polyethylene- glycol (mPEG).
[00059] As used in this application, except as otherwise expressly provided herein, each of the following terms shall have the meaning set forth below.
[00060] As used herein, the term“about” will be understood by one of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term“about” is meant to encompass variations of ±20 % or ±10 %, including ±5 %, ±1 %, and ±0.1 % from the specified value, as such variations are appropriate to perform the disclosed methods. [00061] The term“lower alkyl", when used alone or in combination with other groups, means a straight chained or branched lower alkyl group containing from 1 -6 carbon atoms, preferably from 1-5, more preferably from 1-4 and especially preferably 1- 3 or 1-2 carbon atoms. These groups include, in some embodiments, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, amyl, isoamyl, neopentyl, 1 -ethylpropyl, hexyl, and the like. The lower alkyl moiety of the "lower alkoxy", the "lower alkoxycarbonyl", the "lower akylaminocarbonyT, "lower hydroxyalkyl' and of the "tri-lower alkylsilyT groups has the same meaning as "lower alkyl” defined above.
[00062] The "lower alkenyl” groups are defined as CVG, alkenyl groups which may be straight chained or branched and may be in the Z or E form. Such groups include vinyl, propenyl, 1 -butenyl, isobutenyl, 2-butenyl, 1 -pentenyl, (Z)-2- pentenyl, (E)-2- pentenyl, (Z)-4-methyl-2-pentenyl, (E)-4-methyl-2-pentenyl, pentadienyl, e.g., 1 , 3 or 2,4-pentadienyl, and the like. In some embodiments, the C2-C6- alkenyl groups are C2-C5- , C2-C4-alkenyl groups. In other embodiments, the C2-C6- alkenyl groups are C2-C3- alkenyl groups.
[00063] The term "lower alkynyl” groups refers to C2-C6-alkynyl groups which may be straight chained or branched and include ethynyl, propynyl, 1 -butynyl, 2- butynyl, 1 -pentynyl, 2-pentynyl, 3-methyl-l -pentynyl, 3-pentynyl, 1 -hexynyl, 2- hexynyl, 3-hexynyl and the like. In some embodiments, C2-C6-alkynyl groups are C2-C5-, C2-C4-alkynyl groups. In other embodiments, C2-C6-alkynyl groups are C2-C3-alkynyl groups.
[00064] The term "aryl” group refers to C6-C 14-aryl groups which contain from 6 up to 14 ring carbon atoms. These groups may be mono-, bi- or tricyclic and are fused rings. In some embodiments, the aryl groups include phenyl, biphenyl, naphthyl, anthracenyl, phenanthrenyl and the like. The aryl moiety of the "arylcarbonyl" and the "arylaminocarbonyl" groups has the same meaning as defined above.
[00065] The term "heteroaryl” groups may contain 1 to 3 heteroatoms independently selected from nitrogen, sulfur or oxygen and refers C3-C 13-heteroaryl groups. These groups may be mono-, bi- or tricyclic. In some embodiments, the C3- C heteroaryl groups include heteroaromatics and saturated and partially saturated heterocyclic groups. These heterocyclics may be monocyclic, bicyclic, tricyclic. In some embodiments, the 5 or 6-membered heterocyclic groups are thienyl, furyl, pyrrolyl, pyridyl, pyranyl, morpholinyl, pyrazinyl, methyl pyrrolyl, and pyridazinyl. The C3-C13- heteroaryl may be a bicyclic heterocyclic group. In some embodiments, the bicycbc heterocyclic groups are benzofuryl, benzothienyl, indolyl, imidazolyl, and pyrimidinyl. In some embodiments, the C3-Ci3-heteroaryls are furyl and pyridyl.
[00066] The term "lower alkoxy" includes alkoxy groups containing from 1 to 6 carbon atoms, in some embodiments from 1 to 5, in other embodiments from 1 -4 and in yet other embodiments 1 to 3 or 1 to 2 carbon atoms and may be straight chained or branched. These groups include methoxy, ethoxy, propoxy, butoxy, isopropoxy, tert- butoxy, pentoxy, hexoxy and the like.
[00067] The term "acyl” includes lower alkanoyl containing 1 to 6 carbon atoms, in some embodiments from 1 to 5, from 1 to 4, from 1 to 3 or from 1 to 2 carbon atoms and may be straight chained or branched. These groups include, in some embodiments, formyl, acetyl, propionyl, butyryl, isobutyryl, tertiary butyryl, pentanoyl and hexanoyl. The acyl moiety of the "acyloxy" group has the same meaning as defined above.
[00068] The term "halogen" includes fluoro, chloro, bromo, iodio, and the like.
[00069] The term "aralkyl’ group refers CVCi 5-aralkyl wherein the alkyl group is substituted by an aryl. The alkyl group and aryl may be selected from the Ci-C6 alkyl groups and the C6-C 14-aryl groups as defined above, wherein the total number of carbon atoms is between 7 and 15. In some embodiments the CVC 15-aralkyl groups are benzyl, phenylethyl, phenylpropyl, phenylisopropyl, phenylbutyl, diphenylmethyl, 1 , 1 - diphenylethyl, 1 ,2-diphenylethyl. The aralkyl moiety of the "aralkyloxy" groups has the same meaning as defined above.
[00070] The substituted lower alkyl, alkenyl and alkynyl groups have 1 to 3 independently selected substituents, such as lower alkyl, hydroxy, lower alkoxy, carboxyl, lower alkoxycarbonyl, nitro, halogen, amino, mono- or di- lower alkylamino, dioxolane, dioxane, dithiolane, and dithione. The lower alkyl substituent moiety of the substituted lower alkyl, alkenyl and alkynyl groups, and the lower alkyl moiety of the lower alkoxy, the lower alkoxycarbonyl, and the mono- or di-lower alkylamino substituents of the substituted lower alkyl, alkenyl and alkynyl groups have the same meaning as "lower alkyl” defined above.
[00071] The substituted aryl, the substituted heteroaryl and the substituted aralkyl groups each has 1 to 3 independently selected substituents, such as lower alkyl, hydroxy, lower alkoxy, carboxy, lower alkoxycarbonyl, nitro, amino, mono- or di-lower alkylamino, and halogen. The lower alkyl moiety of the lower alkyl, the lower alkoxy, the lower alkoxycarbonyl, and the mono- or di- lower alkylamino groups among the substituents has the same meaning as Tower alkyl’ defined above.
[00072] The heterocyclic group formed by R5 and R6 combined with a nitrogen atom includes pyrrolidinyl, piperidinyl, piperidino, morpholinyl, morpholino, thiomorpholino, N-methylpiperazinyl, indolyl, and isoindolyl.
[00073] In some embodiments, R1 and R2 are independently selected from the group consisting of hydrogen, halogen, nitro, -CH2OH, -(CH2)kR14, -CH=CH(CH2)mR16, - CºC(CH2)nR15, -CO(CH2)jR4 wherein R4 is -SR7, CH20-(substituted or unsubstituted) lower alkyl (wherein the substituted lower alkyl is in some embodiments methoxymethyl, methoxyethyl or ethoxymethyl), -NR5R6. In some embodiments, each of R1 and R2 is hydrogen.
[00074] In some embodiments of R1 and R2, the residue R14 is selected from phenyl, pyridyl, imidazolyl, thiazolyl, tetrazolyl, -COOR15, -OR15 (wherein R15 is in some embodiments selected from hydrogen, methyl, ethyl, phenyl or acyl), -SR7 (wherein R7 is in some embodiments selected from substituted or unsubstituted lower alkyl, 2-thiazoline and pyridyl) and -NR5R6(wherein R5 and R6 are in some embodiments selected from hydrogen, methyl, ethyl, phenyl, carbamoyl and lower alkylaminocarbonyl). Moreover, in some embodiments, the residue R16 is selected from hydrogen, methyl, ethyl, phenyl, imidazole, thiazole, tetrazole, -COOR15, -OR15 and -NR5R6 (wherein the residues R15, R5 and R6 have the meanings as described above). In some embodiments of R1 and R2, the residue R7 is selected from the group consisting of substituted or unsubstituted lower alkyl, substituted or unsubstituted phenyl, pyridyl, pyrimidinyl, thiazole and tetrazole. Further, in some embodiments, k is 2, 3 or 4, j is 1 or 2 and m and n are independently 0 or 1.
[00075] In some embodiments, R3 is hydrogen or acetyl. Furthermore, in some embodiments, each W1 and W2 is hydrogen.
[00076] In some embodiments, when Y' is a polymer moiety and X' is not a polymer moiety, X' is selected from carboxy, hydroxymethyl or a lower alkoxycarbonyl. In some embodiments X' is selected from methoxy carbonyl.
[00077] In some embodiments, when X' is a polymer moiety and Y' is not a polymer moiety, Y' is selected from hydroxy or acetyloxy. [00078] In some embodiments, the warhead of the polymer conjugate is a derivative of K252a, which has the formula:
Figure imgf000014_0001
[00079] In some embodiments, the polymer conjugate is SNA- 125 (also referred to as CT340), wherein the composition has the formula:
Figure imgf000014_0002
SNA-125 (also referred to as CT340)
[00080] The formulas depicted herein are not limited to any particular stereochemistry, and all stereoisomers and enantiomers thereof are included in this disclosure.
[00081] As described above, several embodiments disclosed herein provide reduced or minimized exposure (e.g., entry into and/or longevity in a non-target site such as the systemic circulation and/or lymphatic system). In some embodiments, exposure at a non-target site is less than 90%, 75%, 50%, 25%, 15%, 10%, 5% or 2% (or less) of the polymer conjugate as compared to a similar active entity that has not been produced according to the embodiments described herein. In some embodiments, desirable rate of clearance from the non-target site (e.g., systemic circulation and/or lymphatic system) for the compositions described herein is increased by at least 10%, 25%, 50%, or 75% or more as compared to non-conjugated controls. As an example, a PEGylated active agent described herein not only penetrates the desired membranes to reach a desired target, but has reduced non-target exposure by at least 20-80% or more as compared to the non- PEGylated active agent. In some embodiments, blood concentrations measured post administration of the compositions described herein are less than about 0.1 ng/ml, less than 1 ng/ml, or less than 10 ng/ml after, e.g., 15 minutes, 30 minutes, 1 hour, 6 hours or 12 hours.
[00082] In some embodiments, reduced exposure at non-target sites contributes to enhanced efficacy. Efficacy may be enhanced because lower concentrations/amounts/dosing schedules are required to achieve the same or similar therapeutic efficacy at the target site (because, for example, the active ingredient stays at the desired target site for a longer time). In one embodiment, concentrations/amounts/dosing schedules are reduced by 25%-75% or more.
[00083] More rapid clearance rates of the active agent once in the non-target site(s) (such as systemic circulation and/or lymphatic system) are also beneficial because this may allow for a higher concentration or more doses to be delivered. This is especially beneficial for active agents in which a subject would benefit from a higher dose but cannot tolerate the higher dose due to toxicity at the non-target site (e.g., systemic toxicity). Faster clearance rates would permit the desired higher dose to be delivered according to the desired schedule. For example, a subject may be able to tolerate daily doses rather than weekly doses because of the reduced exposure.
[00084] In some embodiments, the active agents of the compositions described herein (e.g., indolocarbazole compounds conjugated e.g., with PEG or other polymers) are measured in non-target sites (e.g., the systemic circulation and/or lymphatic system) at less than amounts found when the active agent is delivered without conjugation (e.g., less than 0.5%, 1% or 2% after 6 or 12 hours, as compared with 3-15% (e.g., 3-6%) when the active agent is delivered without conjugation). In some embodiments, the active agents of the compositions described herein (e.g., indolocarbazole compounds conjugated e.g., with PEG or other polymers) are measured in non-target sites (e.g., the systemic circulation and/or lymphatic system) at less than 0.5%, 1% or 2% after 3-24 hours, as compared to an amount 2-20 times greater when the active agent is delivered without conjugation.
[00085] In some embodiments, clearance of the compositions (e.g., the conjugated polymer compounds) occurs within minutes of exposure to the non-target site (e.g., systemic circulation and/or lymphatic system), as opposed to hours. In other embodiments, 50% clearance of the conjugated polymer compounds occurs in less than 5 minutes, 15 minutes, 30 minutes, 1 hour, 6 hours, and 12 hours of exposure to the systemic circulation and/or lymphatic system. Clearance times of the conjugated polymer compounds are reduced by more than 25%, 50%, 75% and 90%, as compared to the non- conjugated active agents or other formulations. These reduced clearance times are beneficial to reduce toxicity and undesired side effects.
[00086] In some embodiments, an active agent may be increasingly toxic as it is metabolized in the non-target site (e.g., systemic circulation and/or lymphatic system) because the metabolites exhibit more toxicity than the original agent. Thus, faster clearance rates, in some cases even before the toxic metabolites are created, are especially beneficial.
[00087] The term“active entity” or“active agent” as used herein should not be understood as limiting the participation of the polymer itself and/or the chemical linking moiety between the polymer and the warhead in defining the pharmacology of the polymer conjugate. In some embodiments, the polymer influences the selectivity and/or inhibitory activity of the polymer conjugate. In some embodiments, the chemical linking moiety between the polymer and warhead influences the selectivity and/or inhibitory activity of the polymer conjugate. In some embodiments, the polymer conjugates exhibit no change in selectivity or inhibitory activity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the polymer conjugates exhibit a significant increase in selectivity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the polymer conjugates exhibit a significant increase in inhibitory activity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the polymer conjugates exhibit a significant increase in selectivity and inhibitory activity against the therapeutic target in comparison with the unconjugated active agent. In some embodiments, the increased selectivity and/or inhibitory activity of the polymer conjugate against the therapeutic target in comparison with the unconjugated active agent causes decrease in undesired biological effects. In some embodiments, the increased selectivity of the polymer conjugate is caused by an increase of the hydrodynamic volume resulting from the conjugated polymer chain. In some embodiments, the polymer chain creates a higher steric hindrance which allows discrimination among the diverse shapes and sizes of the binding sites of different proteins, thus improving selectivity with respect to the active agent alone.
[00088] In several embodiments, various inflammatory skin diseases are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1). The inflammatory skin disease comprises, in some embodiments, psoriasis, psoriasis guttata, inverse psoriasis, pustular psoriasis, psoriatic erythroderma, pruritis associated with any of the various forms of psoriasis, acute febrile neutrophilic dermatosis, eczema, xerotic eczema, dyshidrotic eczema, vesicular palmar eczema, acne vulgaris, atopic dermatitis, contact dermatitis, allergic contact dermatitis, dermatomyositis, exfoliative dermatitis, hand eczema, pompholyx, keloids, rosacea, rosacea due to sarcoidosis, rosacea due to scleroderma, rosacea due to Sweet syndrome, rosacea due to systemic lupus erythematosus, rosacea due to urticaria, rosacea due to herpetic pain, Sweet's disease, neutrophilic hydrodenitis, sterile pustule, drug rash, seborrheic dermatitis, pityriasis rosea, Kikuchi's disease of the skin, pruritic urticarial papules and plaques of pregnancy, Stevens-Johnson syndrome and toxic epidermal necrolysis, tattoo reaction, Wells syndrome (eosinophilic cellulitis), reactive arthritis (Reiter syndrome), bowel-associated dermatosis-arthritis syndrome, rheumatoid neutrophilic dermatosis, neutrophilic eccrine hidradenitis, neutrophilic skin disease of dorsum of hand, balanitis circumscripta plasmacellularis, balanoposthitis, Behcet's disease, erythema annulare centrifugum, erythema dyschromicum perstans, erythema multiforme, granuloma annulare, dermatitis of hand, lichen nitidus, lichen planus, lichen sclerosus et atrophicus, lichen simplex chronicus, lichen spinulosus, nummular dermatitis, pyoderma gangrenosum, sarcoidosis, subcorneal pustular dermatosis, urticaria, transient acantholytic dermatosis, vitiligo, prurigo nodularis, and dry skin. [00089] In some embodiments, provided herein are methods of treating psoriasis (e.g., psoriatic plaques) in a subject in need thereof, comprising administering an effective amount of a formulation provided in Table 1 to the subject.
[00090] In some embodiments, provided herein are methods of treating atopic dermatitis in a subject in need thereof, comprising administering an effective amount of a formulation provided in Table 1 to the subject.
[00091] In some embodiments, provided herein are methods of treating pruritis (e.g., pruritis associated with psoriasis or atopic dermatitis) in a subject in need thereof, comprising administering an effective amount of a formulation provided in Table 1 to the subject.
[00092] In some embodiments, provided herein are methods of treating psoriasis (e.g., psoriatic plaques) in a subject in need thereof, comprising administering an effective amount of a formulation comprising about 0.2 % to about 2.0 % w/w (e.g., about 0.2 %, about 0.5 %, about 1.0 %, or about 2.0 %) of SNA-125 to the subject.
[00093] In some embodiments, provided herein are methods of treating atopic dermatitis in a subject in need thereof, comprising administering an effective amount of a formulation comprising about 0.2 % to about 2.0 % w/w (e.g., about 0.2 %, about 0.5 %, about 1.0 %, or about 2.0 %) of SNA-125 to the subject.
[00094] In some embodiments, provided herein are methods of treating pruritis (e.g., pruritis associated with psoriasis or atopic dermatitis) in a subject in need thereof, comprising administering an effective amount of a formulation comprising about 0.2 % to about 2.0 % w/w (e.g., about 0.2 %, about 0.5 %, about 1.0 %, or about 2.0 %) of SNA- 125 to the subject.
[00095] In some embodiments, the administration is once daily (e.g., once daily for 8 weeks or 12 weeks). In some embodiments, the administration is twice daily (e.g., twice daily for 8 weeks or 12 weeks).
[00096] In some embodiments, the subject has a weekly mean I-NRS score of at least 5 on the 11 point I-NRS scale.
[00097] In some embodiments, the subject has a weekly mean I-NRS score of at least 5 on the 11 point I-NRS scale and the psoriasis is mild-to-moderate psoriasis (e.g., mild-to-moderate plaque psoriasis).
[00098] In some embodiments, the subject has mild-to-moderate psoriasis with at least moderate itch. [00099] In some embodiments, the psoriasis is mild to moderate psoriasis.
[000100] In some embodiments, the psoriasis is mild to moderate plaque psoriasis.
[000101] In several embodiments, various skin neoplasias are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1). The skin neoplasia comprises, in some embodiments, squamous cell carcinoma, basal cell carcinoma, malignant melanoma, malignant cutaneous lymphoma, Kaposi's sarcoma, Merkel cell skin cancer, and non-melanoma skin cancer.
[000102] In several embodiments, various vascular tumors are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1). The vascular tumor comprises, in some embodiments, hemangiomas, Kaposi's sarcoma, lymphangioma, glomangioma, angiosarcoma, hemangioendothelioma, and infantile hemangiomas.
[000103] In several embodiments, various bullous diseases are treated (e.g., treated by administering an effective amount of a formulation provided in Table 1). The bullous disease comprises, in some embodiments, bullous pemphigoid, erythema multiforme, dermatitis herpetiformis, epidermolysis bullosa acquisita, linear Immunoglobulin A disease, mucous membrane pemphigoid, pemphigoid gestationis, pemphigus foliaceus, and pemphigus vulgaris.
[000104] In several embodiments, hair growth and cycling are modulated (e.g., modulated by administering an effective amount of a formulation provided in Table 1). In several embodiments, alopecia is treated.
[000105] In several embodiments, the polymer conjugates (e.g., a formulation provided in Table 1) are administered in combination with UV irradiation therapy.
[000106] Also provided herein, in several embodiments, are polymer conjugates wherein the polymer is polyethylene glycol (PEG) or methoxy-polyethylene glycol (m- PEG). In several embodiments, there is provided a pharmaceutical composition comprising or consisting essentially of a polymer conjugate disclosed herein that is formulated for topical administration. In several embodiments, methods of making and using the compositions described herein are provided.
[000107] In several embodiments, the invention comprises a reduced exposure composition comprising at least one active entity linked to at least one polymer, wherein the composition has reduced exposure at a non-target site as compared to the active entity delivered without the polymer. The non-target site comprises the systemic system, the lymphatic system and/or another non-target tissue site in some embodiments.
[000108] In some embodiments, the active entity binds to a kinase. In some embodiments, the active entity binds to Tropomyosin receptor kinase A (TrkA). In some embodiments, the active entity binds to a Janus Kinase (JAK) family member in some embodiments. In some embodiments, the active entity binds to one or more of Janus Kinase 1 (JAK1), Janus Kinase 2 (JAK2), Janus Kinase 3 (JAK3), and/or Tyrosine kinase 2 (TYK2) in some embodiments. In some embodiments, the active entity binds to mitogen-activated protein kinase kinase 2 (MAP2K) and/or mitogen-activated protein kinase kinase 3 (MAP2K3). Such kinase binding by the active entity may be partially or fully inhibitory or not.
[000109] In some embodiments, the polymer used in the reduced exposure compounds comprises polyethylene glycol (PEG) and/or methoxy-polyethylene glycol (m-PEG). In embodiments where the active entity has one or more carboxyl, hydroxyl, amino and/or sulfhydryl groups, the active entity is PEGylated (or conjugated/coupled to another polymer) at one or more of said carboxyl, hydroxyl, amino and/or sulfhydryl groups.
[000110] The reduced exposure compositions described herein are formulated for topical administration in several embodiments. In several embodiments, methods of treating one or more of the following are provided: inflammatory skin disease, vascular tumors, skin neoplasia, bullous diseases, alopecia, wounds, scars, autoimmune disorders, and cancerous or pre-cancerous lesions. Methods for modulating hair growth and cycling are provided in some embodiments.
[000111] In one embodiment, a method of treating any of the above-mentioned skin diseases or any other skin condition in need of treatment, includes: applying, or instructing application of, a topical formulation (e.g., a formulation provided in Table 1) to a skin region, wherein the formulation fully or partially inhibits signaling in the skin region, and thereby treats the skin condition, wherein the formulation comprises: a polymer conjugate of an indolocarbazole compound in an oil-in-water emulsion. The topical formulation applied in accordance with the method may be any of the gel formulations of SNA-125 as more fully described herein.
[000112] In some embodiments, the compositions provided herein (e.g., a formulation provided in Table 1) may be administered via at least two routes of administration, either simultaneously or sequentially according to some embodiments. In one embodiment, the composition is administered via a first (e.g. topical dermal) route to a subject, wherein the subject further receives an additional agent via a second (e.g., non- dermal) route to achieve synergetic effects.
Synthesis of SNA-125 (CT340t
[000113] In some embodiments, the active agent (SNA-125/CT340) may be synthesized as follows:
Step 1 : Hydrolysis to K252b
Figure imgf000021_0001
[000114] 1 molar equivalent of K252a was dissolved in 7.1 vol of THF and the mixture was stirred for at least 30 minutes at ambient temperature (~20°C). A solution of 3 eq. LiOHxH20 in 4 vol of highly purified water (with respect to K252a) was added to the pale yellow solution over ~ 4 minutes to give a biphasic system which was stirred for ~2l h at ambient temperature, after which an IPC test by HPLC indicated <2% a/a K252a.
[000115] The majority of the solvent was distilled off under reduced pressure at ~30°C. 3 vol of highly purified water were added to the residue and evaporation under reduced pressure at ~30°C was continued to remove the remainder of the solvent, resulting in a thick aqueous mixture. The mixture was cooled to ~20°C and 2N HC1- solution (approximately 3-3.5 vol) was added to adjust the pH to 2-3, affording a white suspension. The suspension was stirred for ~40 minutes at ambient temperature and the solid collected by suction filtration, washing twice with highly purified water. The solid was then tumble-dried on the rotary evaporator at ~25°C under reduced pressure. It was then slurried in 14.2 vol of ethyl acetate for ~ 1 h. The solid was collected by suction filtration, washing twice with heptane. The solid was then tumble-dried on the rotary evaporator at - 25°C under reduced pressure, affording K252b in essentially quantitative (uncorrected) yield.
Step 2: Coupling
Figure imgf000022_0001
CJ»
[000116] 1 molar equivalent of K252b was pre-dried by suspending in 10 vol of dichloromethane and concentrating to dryness on the rotary evaporator at ~25°C then holding under reduced pressure to afford a yellowish solid. The dried K252b was re suspended in 160 vol of dichloromethane at ambient temperature (~20°C) and 2 molar equivalents of 4-methylmorpholine were added. The resulting suspension was stirred for ~ 35-45 minutes.
[000117] 1 molar equivalent of PEG-amine was pre-dried by dissolving in dichloromethane (10 vol with respect to K252b) and concentrating to dryness on the rotary evaporator at ~25°C to give a white solid. The dried PEG-amine was dissolved in 50 vol of dichloromethane and this solution was added over ~20 minutes to the K252b/4- methylmorpholine suspension. The reaction mixture was stirred for ~20 minutes at ambient temperature and 1.7 eq. TBTU was then added in one portion. The reaction mixture was stirred for ~l8-22h at ambient temperature, after which an IPC test by HPLC indicated <2% K252b.
[000118] Methanol (0.01 vol) was added to quench the reaction and the mixture was stirred for -1-1.5 hours at ambient temperature. 30 vol of saturated sodium bicarbonate solution were added and the biphasic mixture was then stirred for -20 minutes. The phases were separated and the lower (organic) phase was washed with 15 vol of saturated sodium bicarbonate solution. The organic phase was concentrated to dryness under reduced pressure at - 25°C, protected from light. Step 3: Purification and Isolation
[000119] The product from Step 2 is purified by preparative column chromatography using silica gel which has been pretreated with ethyl acetate containing 5% triethylamine. The Step 2 product was dissolved in dichloromethane/ethyl acetate for application to the column. Elution with ethyl acetate enabled residual K252a and UV- inactive species to be eluted (as monitored by TLC). The column was then conditioned with dichloromethane and eluted with dichloromethane/MeOH (98.5: 1.5 - 95.5:4.5). Fractions were collected and analyzed by HPLC and TLC, and pooled according to their purity. Evaporation of the combined desired product fractions led to CT340 of purity >95% by HPLC area.
[000120] Although the resulting material is of high chemical purity, a precipitation step is performed to transform it from a semisolid concentrate into a readily handled free-flowing solid.
[000121] The concentrate was therefore dissolved in 4 vol of MeOH and this solution was added to chilled (~0°C) diethyl ether (20 vol) over 40-70 minutes to afford a white suspension that was stirred for further 1-2 hours at ~0°C. The solid product was filtered off, washing with diethyl ether (2 x 2 vol), to give CT340 Drug Substance in a typical yield of - 90%.
Formulations for topical delivery
[000122] In some embodiments, the active agent is formulated for topical delivery. The active agent may be a reduced exposure composition. In certain embodiments, the active agent is a polymer conjugate of an indolocarbazole compound, thereby providing a reduced exposure indolocarbazole compound. In one particular embodiment, the polymer conjugate is the indolocabazole depicted in Formula (II). In another particular embodiment, the polymer conjugate is SNA-125 (formerly referred to as CT340; both terms are used interchangeably herein). The topical delivery formulation can be in any form, including for example, an ointment, a gel, a balm, a cream, a lotion, a primer, a serum, a liquid, a spray, etc. In some embodiments, the topical delivery formulation is a gel.
[000123] The weight percentage (w/w) of active agent to the total weight of the topical delivery formulation may range from about 0.001% to about 20%, from about 0.005% to about 15%, from about 0.01% to about 12%, or from about 0.1% to about 10%, including any weight percentages within the disclosed ranges. In other embodiments, the weight percentage (w/w) of the active agent may be greater than or equal to about 1.00% w/w. In certain embodiments, the weight percentage (w/w) of the active agent to the total weight of the topical delivery formulation may fall within any range defined by any two of the above weight percentages.
[000124] In other embodiments, the weight percentage (w/w) of the active agent may be less than or equal to about 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% w/w (e.g., less than or equal to about 5.0%, 2.0%, 1.0%, 0.5%, or 0.2%). In certain embodiments, the weight percentage (w/w) of the active agent to the total weight of the topical delivery formulation may fall within any range defined by any two of the above weight percentages.
[000125] In certain embodiments of the topical delivery formulation, the active agent is a polymer conjugate of an indolocarbazole compound providing a reduced exposure. In one particular embodiment, the polymer conjugate is the indolocabazole depicted in Formula (II). In another particular embodiment, the polymer conjugate is SNA- 125.
Gel formulation
[000126] In one embodiment, a gel formulation includes: a polymer conjugate of an indolocarbazole compound, a preservative/solvent, a solvent, a penetration enhancer, a pH adjusting agent, a gelling agent, and a vehicle. In an embodiment of the gel formulation, the polymer conjugate of an indolocarbazole compound is SNA-125. The SNA-125 may be present at about 0.05% to about 25% (w/w) of the gel formulation. In a variation, the SNA-125 may be present at about 0.5% to about 10% (w/w) of the gel formulation. In another variation, the SNA-125 may be present at about 5% (w/w) of the gel formulation.
[000127] In certain embodiments of the disclosed gel formulation, the preservative/solvent may be one or more selected from benzoic acid, sorbic acid, boric acid, methylparaben, ethylparaben, propylparaben, butylparaben, sodium benzoate, sodium propionate, potassium sorbate, chlorobutanol, benzyl alcohol and phenyl ethyl alcohol, phenol, chlorocresol, o-phenyl phenol, benzalkonium chloride, cetyl pyridinium chloride, imidurea, thimerisal, propyl gallate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), citric acid, ascorbic acid, ascorbyl palmitate, alpha-tocopherol and and disodium edetate. In one embodiment, the preservative/solvent is benzyl alcohol. The preservative/solvent may be present at about 0.1% to about 10% (w/w) of the gel formulation, including any concentrations defined by the range. In a variation, the preservative/solvent may be present at about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, or 10% (w/w), and may fall within any range defined by any two of the above weight percentages. Alternatively, the preservative/solvent may be present at about 1% (w/w) of the gel formulation.
[000128] In certain embodiments of the disclosed gel formulation, the solvent may be one or more selected from ethanol, propylene glycol, glycerin and polyethylene glycol. In one embodiment, the solvent is propylene glycol. The solvent may be present at about 1% to about 30% (w/w) of the gel formulation, including any concentrations defined by the range. In a variation, the solvent may be present at about 1%, 5%, 10%, 15%, 20%, 25% or 30% (w/w), and may fall within any range defined by any two of the above weight percentages. Alternatively, the solvent may be present at about 20% (w/w) of the gel formulation.
[000129] In certain embodiments of the disclosed gel formulation, the penetration enhancer may be one or more selected from dimethylsulfoxide, Transcutol, oleic acid, azone, pyrrolidones, surfactants (e.g. SLS, Tween), urea, essential oils, terpenes and terpenoids, and phospholipids. In one embodiment, the penetration enhancer is dimethylsulfoxide (‘DMSO’). The penetration enhancer may be present at about 1% to about 30% (w/w) of the gel formulation, including any concentrations defined by the range. In a variation, the solvent may be present at about 1%, 5%, 10%, 15%, 20%, 25% or 30% (w/w), and may fall within any range defined by any two of the above weight percentages. Alternatively, the penetration enhancer may be present at about 20% (w/w) of the gel formulation.
[000130] In certain embodiments of the disclosed gel formulation, the pH adjusting agent may be one or more selected from sodium hydroxide, hydrochloric acid, phosphoric acid, and triethylamine. In one embodiment, the pH adjusting agent is trolamine. The pH adjusting agent may be used if necessary in an amount sufficient to adjust the final pH of the gel formulation (‘q.s.’) to greater than or equal to about 5.0. In a variation, the final pH may be adjusted (‘q.s.’) to between about 5.0 and about 7.5, or to any pH selected from about 5, 5.2, 5.4, 5.6, 5.8, 6.0, 6.2, 6.4, 6.6, 6.8, 7.0, 7.2, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0.
[000131] In certain embodiments of the disclosed gel formulation, the gelling agent may be one or more selected from hydroxyethyl cellulose, hydroxypropyl methylcellulose, carbopol, gelatin, sodium alginate, sodium carboxymethylcellulose, gelatin, polysaccharides, and silica. In one embodiment, the gelling agent is hydroxyethyl cellulose (‘HEC’)· The gelling agent may be present at about 0.05% to about 20% (w/w) of the gel formulation. Alternatively, the gelling agent may be present at about 0.5% to about 5% (w/w), including any concentrations defined by the range. In a variation, the gelling agent may be present at about 0.50%, 0.75%, 1.0%, 1.25%, 1.5%, 1.75%, 2.0%, 2.25%, 2.5%, 2.75%, 3.0%, 3.25%, 3.5%, 3.75%, 4.0%, 4.25%, 4.5%, 4.75% or 5.0% (w/w), and may fall within any range defined by any two of the above weight percentages. Alternatively, the gelling agent may be present at about 1.25% (w/w) of the gel formulation.
[000132] In certain embodiments of the disclosed gel formulation, the vehicle may be an aqueous solvent, such as purified water, which is used in an amount sufficient to bring the gel formulation to 100% (w/w) - i.e., purified water q.s.
[000133] In some embodiments, the gel formulation is one of the gel formulations of Table 1. In one embodiment, a gel formulation for dermal delivery of SNA-125 with reduced exposure is disclosed, wherein the gel formulation includes about 5% w/w SNA-125, about 1% w/w benzyl alcohol, about 20% w/w/ propylene glycol, about 20% w/w DMSO, and about 1.25% w/w HEC, where trolamine is added q.s. to adjust the pH to between about 5.0 and about 7.5, and purified water q.s. An embodiment of such an SNA- 125 gel formulation is set forth above in Table 1.
EXAMPLE 1: CT340 FORMULATION DEVELOPMENT
Summary
[000134] Initial formulation development of a topical drug product containing CT340 (SNA-125) at 5% w/w for the local treatment of pain was undertaken. The overall objective was to develop a chemically and physically stable topical product which could be stored at room temperature. All considered excipients were selected from FDA’s Inactive Ingredients database. Further, the drug had to be in a dissolved form and suspensions were avoided.
[000135] During early stages of formula development, several product and formulation concepts were considered. An aqueous gel was most preferred, but other dosage forms were explored as well. Consequently the preformulation activities encompassed evaluating the solubility and compatibility of the drug substance in a broad spectrum of solvents suitable for use in these dosage forms. The initial goal was to investigate the stability of CT340 in solubilized (dissolved) form in various solvent blends. Data generated during this preliminary feasibility evaluation was utilized to develop three different formulation approaches, namely a) multiple aqueous gels, b) a petrolatum based ointment and c) a lipophilic solution composition.
[000136] This Example summarizes the development history, including formulation design and preliminary stability of the various formulation prototypes.
Investigation of the Solubility and Compatibility of CT340 in Solvent Blends
Purpose
[000137] The purpose of this solubility study was to identify appropriate solvents and/or solvent blends suitable for the development of diverse dosage forms. The target dosage forms were aqueous gels, lipophilic gels and petrolatum-based ointments. Only solvents present in the FDA's Inactive Ingredients Guide (IIG) for approved topical products were considered. Solubility was assessed by visual determination.
Methodology
[000138] A small amount of API (-0.25 g) was weighed into type 1 clear glass vials. To each vial, approximately 4.75 g of the solvent or solvent blend was added. Samples were shaken manually and visually observed to check if the API had dissolved. Clear solution indicated that the drug had dissolved. The resultant solutions were stored at 5°C and 40°C for two weeks. These samples were evaluated for their color, the clarity of the solution and assayed for CT340 content.
Results and Conclusions
[000139] Results of the compatibility testing are provided in Table 2 below. Data from the experiments indicate that the drug can be dissolved at 5% w/w in a variety of solvent blends, permitting the development of diverse dosage forms. All solutions were light yellow in color initially. After 2 weeks at 5°C and 40°C, alcoholic solutions had remained unchanged, while the presence of Transcutol resulted in a darker yellow solution at 40°C. Other experimental solvent exhibited a slightly more intense yellow compared to the initial. The drug substance remained dissolved in all blends after two weeks at both 5°C and 40°C temperatures. CT340 appears to be compatible with all solvents tested other than Transcutol.
TABLE 2: SOLUBILITY AND COMPATIBILITY OF 5% W/W CT340 IN
SOLVENTS AND SOLVENT BLENDS
Figure imgf000028_0001
Investigation of Miscibility of Solvents
Composition of Solvent Blends [000140] The drug substance CT340 does not exhibit significant solubility in non-polar solvents alone such as isopropyl myristate and caprylic capric triglyceride. However, it exhibits sufficient solubility in alcohol to make it possible to develop prototypes containing alcohol and non-polar solvents. Examples of these are seen in Table 2 (3650-6A and 6B). While the concentration of alcohol used in those blends is below the maximum listed in the IIG, it was desirable to lower it in the CT340 prototypes to minimize its potential sensory effect on the skin during the clinical trials. This necessitated identifying solvent which could be used as "fillers", which were listed on the IIG at relatively high concentrations, would be mutually miscible when used at the required levels and still keep the drug in solution. Table 3 provides the composition of the two blends evaluated.
TABLE 3: COMPOSITION OF LIPOPHTT JC SOLVENT BLENDS
Figure imgf000029_0001
Conclusions
[000141] Adjustment of the ratios of the polar and non-polar solvents results in a miscible blend of solvents. The blend has the potential balance between solubilizing the drug substance and helping deliver it through the skin. Addition of 5% CT340 to this blend resulted in a clear solution. This composition was expected to form the base of a lipophilic gel prototype.
Selection of Gelling Agents
[000142] Three types of dosage forms were considered: an aqueous gel, a lipophilic gel and an ointment. As described in above section, suitable solvent blends had already been identified. The objective of the work described in this section of the Example was to identify appropriate gelling agents for the aqueous and alcoholic solvent blends.
Aqueous Gels
[000143] The solvent blend for this prototype consisted mainly of purified water, DMSO and propylene glycol. Both hydroxyethylcellulose (HEC) and hydroxypropyl methylcellulose (HPMC) are commonly used to gel blends of water and propylene glycol. Both polymers are also in common use in topical products. However, the effectiveness of these polymers in gelling aqueous systems containing DMSO was unknown. It was therefore decided to test the gelling ability of HEC and HPMC using water and propylene glycol containing DMSO at the highest proposed concentration (40% w/w). The test compositions are provided in Table 4.
TABLE 4: TEST COMPOSITIONS FOR GELLING ABILITY OF HEC AND
HPMC
Figure imgf000030_0001
[000144] Composition 10A was a clear viscous gel while 10B was a 'lumpy' runny product. For this type of base, therefore, HEC was selected for use as the gelling agent.
Lipophilic Gel
[000145] The solvent composition for this prototype consisted of Isopropyl Myristate, Oleic Acid, Caprylic Capric Triglyceride, Alcohol, Propylene Glycol and Benzyl Alcohol.
[000146] It was shown the above section that this balanced blend of polar and non-polar solvents is miscible and able to dissolve 5.0% w/w of CT340. [000147] Two different approaches were evaluated to gel the lipophilic solvent blend: using silica, a mineral gelling agent, and hydroxypropylcellulose (HPC), a cellulosic gelling agent. Silica exhibits good gelling of lipophilic solvents, whereas HPC is very effective at gelling alcohol based vehicles.
[000148] Neither ingredient was found to be effective at suitably gelling the lipophilic solvent blend of interest. Further optimization of the solvent blend is necessary to develop a gel using this solvent blend. For the purpose of assessing the skin- penetration properties of the drug from diverse prototypes, it was decided to proceed with the solution (no gelling agent) and revisit the gelling properties if it was deemed to be the best at delivering CT340 through the skin.
Development of Prototypes
[000149] Based on the information attained from the solubility and compatibility experiments with CT340 and from the experimentation with the gelling agents, five different prototypes (Table 5) were selected for further development work. These may be divided into three broad categories: (a) aqueous gels - formula #s 3650-16B, 3650-17A and 3650-18A; (b) lipophilic solution - formula # 3650-19A; and (c) petrolatum based ointment - formula # 3650-20 A.
[000150] Quantitative compositions of the five prototypes are provided in Table 5. The three aqueous gels are all thickened with hydroxy ethylcellulose. The three differ from each other primarily in their concentrations of DMSO, propylene glycol and benzyl alcohol. The lipophilic solution consists of a blend of polar and non-polar solvents. As mentioned earlier, the fifth prototype is a petrolatum based ointment in which the drug phase is dispersed in the waxy base. The drug phase utilizes benzyl alcohol and propylene glycol to dissolve the drug substance.
Investigation of the Stability of Prototypes
[000151] Prototypes with the compositions described in Table 5 below were packaged in type 1 clear glass vials and tested for stability as described in the Stability Protocol below. TABLE 5: QUANTITATIVE COMPOSITION OF CT340 PROTOTYPES
Figure imgf000032_0001
Stability Protocol
[000152] This stability study is designed to determine the chemical and physical stability of CT340 5.0% w/w prototype formulations. These formulation prototypes were prepared as ~60g (gels) & lOOg (ointment) laboratory batches. All batches there then split to have approximately 5g in scintillation vials for stability study. The prototypes were stored at 25°C/60% RH, and 40°C/75%RH for up to 6 months. The storage conditions and testing schedule are depicted in Tables 6 and 7 below. For the packaging, glass vials were wrapped with aluminum foil. There were nine total vials per formulation. The sample orientation was vertical, and sample storage after pull and prior to testing was controlled room temperature. The following testing was performed: appearance (visual, single determination) and assay (duplicate determination).
TABLE 6: STORAGE CONDITIONS FOR STABILITY STUDY
Figure imgf000033_0002
Figure imgf000033_0001
Figure imgf000033_0003
X -Physical testing
Y ~ Chemical testing
Results
[000153] The stability test results are summarized in Table 8. The results indicate that CT340 is chemically stable in all three aqueous gels and in the lipophilic solution. These four compositions also exhibit acceptable physical stability. The ointment, however, had separated into two phases within a month at both conditions. The significantly higher variability observed in the assay values may be a reflection of this physical instability.
TABLE 8: STABILITY OF CT340 PROTOTYPES
Figure imgf000034_0001
Figure imgf000034_0003
Figure imgf000034_0005
Figure imgf000034_0002
Figure imgf000034_0004
Values in parentheses are the individual assay values from duplicate determination
" Sample exhibited two phases, ointment at the top and liquid at the bottom of the vial.
J 25C samples were evaluated at 3 months; 40C samples were evaluated at 2 months.
NC: No change
NT: Not tested
Laboratory Manufacturing Process
[000154] The laboratory process described in this section focuses on the manufacture of the gels since these prototypes performed the best at delivering CT340 into the dermal tissue and also exhibited acceptable stability. Prototype 3650-17A is taken as an example.
Laboratory Manufacturing Process for Gel Prototype 3650-17A
(1) To the main manufacturing vessel, add Purified Water, Propylene Glycol, DMSO, Benzyl Alcohol and Sodium Laureth Sulfate.
(2) With continuous stirring, add CT340. Mix until dissolved.
(3) If necessary, adjust pH to >5.0, using Trolamine solution. (4) While mixing with a propeller mixer, slowly add the Hydroxy ethylcellulose.
(5) Mix until the polymer is dissolved and a clear, yellowish gel is attained.
(6) Fill into the appropriate container-closure system.
Summary
[000155] Based on a brief assessment of solubility and compatibility of CT340 in pharmaceutically acceptable ingredients, five prototypes were selected for development work. Three of these were aqueous gels, one was a lipophilic solution and the fifth was an ointment. Four of the five prototypes have exhibited acceptable physical and chemical stability. The ointment was the sole exception, as it exhibited phase separation within a month. All five compositions were also evaluated in an in vitro skin penetration study described in Example 2. As described in Example 2, the aqueous gel formula 3650-17A, 18A and 19A performed the best of the five compositions evaluated. Taking into consideration, the physical and chemical stability of the various prototypes, their skin penetration performance and suitability for the desired indication, the aqueous gel formula, 3650-17A has been selected for further development.
EXAMPLE 2: IN VITRO PERCUTANEOUS ABSORPTION OF CT340 FROM
PROTOTYPE FORMULATIONS USING HUMAN TISSUE
Summary
[000156] Drug product efficacy is determined by controlled clinical trials. Bioavailability and potency of topically applied active pharmaceutical ingredients are key components in efficacious drug products. Potential cutaneous (local) and/or systemic bioavailability can be assessed using in vitro percutaneous absorption testing. Data generated using in vitro skin permeation models can support formulation selection during pharmaceutical development programs.
[000157] Topical formulations containing CT340 are under development for the treatment of pain. The purpose of this study was to characterize in vitro percutaneous absorption of CT340 from topical formulations following application to excised human skin from elective surgery.
[000158] This study was conducted using procedures adapted from the FDA and AAPS Report of the Workshop on Principles and Practices of In Vitro Percutaneous Penetration Studies: Relevance to Bioavailability and Bioequivalence (Skelly et al, 1987). Human tissue from a single donor was dosed with 5 mg/cm2 of formulation.
[000159] Data from this preliminary in vitro skin permeation experiment indicated that prototype CT340GP3 (G) exhibited the highest efficiency of delivery for CT340 as well as the highest tissue deposition of CT340.
[000160] After statistical evaluation, deposition profiles of CT340 into the epidermis and dermis can be rank ordered as follows: CT340GP3 (G) = CT340GP2 (F) = CT340AhGP4 (H) > CT340GP1 (E) > CT3400P5 (I).
[000161] Numerically, CT340GP3 (G) is the most efficient at delivering CT340 into the tissue layers while CT3400P5 (I) is the least efficient.
[000162] In addition to the delivery potential of the prototypes, final prototype selection is expected to factor in other evaluation parameters such as physical and chemical stability of the formulations, disease state and availability of supporting toxicological data.
Introduction
[000163] The purpose of this study was to characterize in vitro percutaneous absorption of CT340 from prototype formulations following application to excised human skin from elective surgery. In vitro skin permeation methodology was utilized to assess the effect of formulation modification on the delivery of CT340 from prototype compositions.
[000164] In vitro skin permeation data is intended to facilitate identification of formulation candidates with the highest potential of success, in terms of developing a topical formulation with appropriate delivery characteristics. This approach may also provide the highest potential for correlating in vitro permeation data with clinical efficacy in diseases where the site of action is located in the viable tissues of the skin.
Methods
[000165] This in vitro percutaneous absorption study was conducted using procedures adapted from the FDA and AAPS Report of the Workshop on Principles and Practices of In Vitro Percutaneous Penetration Studies: Relevance to Bioavailability and Bioequivalence (Skelly et al, 1987). The compositions of the CT340 formulations evaluated in this study are summarized in Table 9. TABLE 9 CT340 PROTOTYPE FORMULATION COMPOSITIONS
Figure imgf000037_0001
’Sodium Inuryi !sulf !te was listed 5» the erigirsa! composition (able found in the signed protocol and changed to So ium htureth sulfate per client request at the time of compounding.
[000166] The clinically relevant dose of 5 mg/cm2 of formulation was applied to dermatomed human abdominal tissue from a single donor obtained following elective surgery. The thickness of the tissue ranged from 0.022 - 0.033 inches (0.559 - 0.838 mm) with a mean +/- standard deviation in thickness of 0.028 +/- 0.003 inches (0.708 +/- 0.084 mm) and a coefficient of variation of 11.9%.
[000167] Percutaneous absorption was evaluated using this human abdominal tissue from a single donor mounted in Bronaugh flow-through diffusion cells. The cells were maintained at a constant temperature of 32 °C by use of recirculating water baths. These cells have a nominal diffusion area of 0.64 cm2. Following the 24-hour duration exposure, the formulation residing on the tissue surface was removed by tape-stripping with CuDerm D-Squame stripping discs. These have been retained for potential future analysis. The epidermis, dermis, and receptor phase samples were labeled and frozen prior to subsequent analysis of CT340 content by LC/Fluorescence. [000168] The tissue sample limit of quantification (LOQ) achieved by the R&D LC/Fluorescence analytical method for CT340 was 1.058 ng/mL.
[000169] LC/Fluorescence was selected as the detection technique for this study. LC/MS was investigated, but was less sensitive than the LC/fluorescence technique.
Methodology details are listed below:
Column: Waters XBridge Cl 8, 3.5pm, 4.6 c 75mm
Flow Rate: 0.5 mL/min
MPA: 5% MeOH, 95% H20, 1% glacial acetic acid
MPB: 100% MeOH, 1% glacial acetic acid
Gradient: Isocratic, 30% A, 70% B
Injection Volume: 5pL
Column Temperature: 40°C
Autosampler Temperature: 20°C
Runtime: 5 minutes
Ex: 335 nm
Em: 400 nm
[000170] Tissue permeation and deposition results were statistically evaluated using unpaired student's t-tests (significant differences between formulations were defined by a p-value of < 0.05, at the 95% confidence interval). Significant differences between formulations were defined by a p value of < 0.05. Outlier tests were performed using Grubbs Test. Outliers in the tissue data whether detected in the epidermis data or in the dermis data were removed from the analysis of both tissue types.
Results
The results of dermal and epidermal deposition are summarized in Table 10.
TABLE 10: CUMULATIVE TISSUE LEVELS OF CT340 FOLLOWING 24
HOURS OF TOPICAL EXPOSURE
Figure imgf000039_0001
Efficiency of Delivery
[000171] The efficiency of delivery is characterized by expressing amount of drug permeating as a percent of applied dose.
Dermal deposition
[000172] Dermal deposition of CT340 from the evaluated formulations ranged from 2.75 to 8.41 percent of the applied dose. Formulations CT340AhGP4 (H) and CT340GP3 (G) had the highest efficiency of CT340 dermal deposition with 6.59 and 8.41 percent of the applied dose, respectively. Overall, formulation CT3400P5 (I) exhibited the least efficient delivery into the dermis.
Epidermal deposition
[000173] Epidermal deposition of CT340 from the evaluated formulations ranged from 10.1 to 17.2 percent of the applied dose. Formulations CT340GP2 (F) and CT340GP3 (G) had the highest efficiency of CT340 epidermal deposition with 16.4 and 17.2 percent of the applied dose, respectively. Formulation CT3400P5 (I) exhibited the least efficient delivery into the epidermis.
Total Amount Delivered
Dermal deposition [000174] The calculated CT340 dermal deposition ranged from 6,867 to 21,034 ng/cm2. Formulations CT340AhGP4 (H) and CT340GP3 (G) generated the highest CT340 dermal deposition with 16,472 and 21,034 ng/cm2, respectively. Formulation CT3400P5 (I) produced the lowest CT340 dermal deposition, 6,867 ng/cm2.
[000175] Dermal levels of CT340 following 24 hour duration of topical exposure are presented in Figure 1 as “calculated nanograms of active substance delivered per square centimeter of skin surface area (ng/cm2).”
Epidermal deposition
[000176] The calculated CT340 epidermal deposition ranged from 25,341 to 43,024 ng/cm2. Formulations CT340GP2 (F) and CT340GP3 (G) had the highest CT340 epidermal deposition with 41,119 and 43,024 ng/cm2, respectively. Formulation CT3400P5 (I) generated the lowest CT340 epidermal deposition, 25,341 ng/cm2.
[000177] Epidermal levels of CT340 following 24 hour duration of topical exposure are summarized in Figure 2 as“calculated nanograms of active substance delivered per square centimeter of skin surface area (ng/cm2).”
[000178] Figure 3 presents the summed dermal and epidermal levels for all formulations used in this study.
Statistical Analysis
[000179] Multiple T-Test analyses (see Tables 11, 12 and 13) were performed, and formulations CT340GP2 (F), CT340GP3 (G), and CT340AhGp4 (H) were found to be equivalent in terms of their combined epidermal and dermal deposition since they were not significantly different from each other and yielded a deposition of CT340 which was significantly different to that of CT340GP1 (E) and CT3400P5 (I).
TABLE 11: T-TEST STATISTICAL ANALYSIS OF DERMAL LEVELS OF
Figure imgf000040_0001
TABLE 12: T-TEST STATISTICAL ANALYSIS OF EPIDERMAL LEVELS OF
CT340 (NG/CM2!
Figure imgf000041_0001
TABLE 13: T-TEST STATISTICAL ANALYSIS OF COMBINED EPIDERMIS
AND DERMIS LEVELS OF CT340 (NG/CM2!
Figure imgf000041_0002
Conclusions
[000180] The purpose of this study was to characterize in vitro percutaneous absorption of CT340 from prototype formulations following application to excised human skin from elective surgery.
[000181] In vitro skin permeation data is intended to facilitate identification of formulation candidates with the highest potential of success, in terms of developing a topical formulation with appropriate delivery characteristics. This approach may also provide the highest potential for correlating in vitro permeation data with clinical efficacy, in diseases where the site of action is located in the viable tissues of the skin.
[000182] Data from this preliminary in vitro skin permeation experiment indicated that prototype CT340GP3 (G) exhibited the highest efficiency of delivery for CT340 as well as the highest tissue deposition of CT340.
[000183] CT340 formulations CT340GP2 (F), CT340GP3 (G) and CT340AhGP4 (H) are not significantly different from each other and show similar deposition profiles; thus, we can consider them equivalent for the purpose of rank ordering. It should be noted that CT340GP2 (F), CT340GP3 (G) and CT340AhGP4 (H) are each significantly different to Formulations CT340GPa (E) and CT3400P5 (I). Therefore, deposition profiles of CT340 into the epidermis and dermis can be rank ordered as follows: CT340GP3 (G) = CT340GP2 (F) = CT340AhGP4 (H) > CT340GP1 (E) > CT3400P5 (I). Overall, CT340GP3 (G) is numerically the most efficient at delivering CT340 into the tissue layers while CT3400P5 (I) is the least efficient.
[000184] In addition to the delivery potential of the prototypes, final prototype selection is expected to factor in other evaluation parameters such as physical and chemical stability of the formulations, disease state and availability of supporting toxicological data.
EXAMPLE 3: EVALUATION OF THE SAFETY AND EFFICACY OF TOPICALLY APPLIED SNA-125 GEL IN HEALTHY VOLUNTEERS AND
SUBJECTS WITH ATOPIC DERMATITIS IN A DOUBLE-BLIND.
RANDOMIZED. INTRAINDIVIDUAL. VEHICLE-CONTROLLED STUDY
[000185] The study subjects consisted of 2 sequential cohorts of adult male or female subjects: (1) healthy volunteers (HV) and (2) subjects with atopic dermatitis (AD). In Cohort 1, 4 HV were evaluated for the safety of topically applied SNA-125 gel and vehicle following 7 days of treatment. The gel formulations (see Table 1) contain SNA- 125 (active ingredient), benzyl alcohol, propylene glycol, dimethylsulfoxide (DMSO), hydroxyethylcellulose (HEC), trolamine, and water (as needed) (the vehicle formulation was the same as the SNA-125 formulation without the active). In Cohort 2, 30 subjects with AD were included in a double-blind, randomized, intraindividual, 4-arm, vehicle- controlled design to evaluate the safety and efficacy of topically applied SNA- 125 gel, applied once-daily for 14 days. Subjects with AD were required to have at least 4 application areas with AD lesions of moderate severity defined by a Total Signs Score (TSS) of >5. Four study products were applied in the clinic without occlusion at approximately 4 mg/cm2 on individual application areas of 3 cm in diameter (approximately 7cm2). Application areas were randomized (1 : 1 : 1 : 1) to SNA-125 0.2 %, SNA-125 2 %, vehicle, and mometasone 0.1 % cream. Six skin biopsies were collected from all AD subjects for immunohistochemistry (IHC) and gene expression analysis using RT-PCR and genomic profiling. Two biopsies were taken at baseline (Day 1), 1 from non-lesional and 1 from lesional skin, and 4 biopsies were taken at Day 15 within application areas (1 biopsy per application area).
[000186] TSS was determined as follows. Six items (erythema, edema/papulation, oozing/crusting, excoriation, lichenification, and dryness) were selected to evaluate the AD severity. The intensity of each item was graded using the following 4-point scale: 0 = absence; 1 = mild; 2 = moderate; and 3 = severe (half steps not allowed). The area chosen for grading was required to have been representative (average intensity) for each item. The individual intensity ratings for each item were then added (ranging from 0-18). The sum of the measures above represented the lesional TSS, which can vary from 0 to 18.
[000187] This study showed that topical application of SNA-125 (0.2 % and 2 %) was well tolerated and showed no safety signals in both healthy subjects and patients with atopic dermatitis, with no serious adverse events reported. Similar modest reductions in clinical scores of target lesions were observed for both SNA- 125 and vehicle.
[000188] Analyses of histology and biomarkers were conducted on biopsy samples obtained from lesional and non-lesional skin from all subjects.
[000189] Effects on various biomarkers related to AD were observed in the SNA-125 0.2 % dose. For example, a reduction in epidermal thickness was observed, as well as changes in the expression levels of certain relevant disease genes. The following genes showed statistically significant expression differences compared to baseline: NGFR (p75), NGF, NTrKl, IL-23/pl9, IL23A, IL 5, IL4R, CCL5, cGRP,CxCL2, CxCLl, DEFB4B, S100A9, S100 A7, CCL26, CCL13, IL32, CxCL9, CxCLlO. As compared to vehicle, expression levels of the following genes were statistically significantly different for SNA- 125 0.2 %: NGFR (p75), TPSB2, IL5, and CCL26.
[000190] For SNA-125 0.2 %, there was a statistically significant correlation between positive improvement in the expression of some relevant disease genes and improvement in clinical TSS score.
EXAMPLE 4: PHASE 1 RANDOMIZED. VEHICLE- AND COMPARATOR- CONTROLLED. DOUBLE-BLIND TRIAL TO ASSESS EFFICACY AND SAFETY OF SNA-125 IN A PSORIASIS PLAQUE TEST
[000191] Psoriasis is a common and often chronic skin disease that affects approximately 2 % of the population. Chronic plaque psoriasis is the most common form, affecting 85 to 95 % of all patients with the disease. The rate of psoriasis varies according to age, gender, region and ethnicity. Chronic plaque psoriasis is characterized by raised, well-demarcated, erythematous oval plaques covered with adherent silvery scales and is most commonly found on the scalp, nails, lower back and the extensor sides of the joints. The scales result from a hyperproliferative epidermis with premature maturation of keratinocytes and incomplete comification, with retention of nuclei in the stratum comeum. This thickening of the epidermis and an inflammatory infiltrate result in thickening of the skin and the raised nature of the skin lesions observed clinically. The inflammatory infiltrate consists of dendritic cells, macrophages and T-cells in the dermis, with neutrophils and some T-cells in the epidermis. Abnormalities of the innate immune system are also involved.
[000192] While systemic biologies have greatly improved the therapeutic options for moderate to severe psoriasis, the majority of psoriasis patients have mild-to- moderate disease. Topical corticosteroids and vitamin D analogs are first-line for treating mild-to-moderate disease and almost 90 % of all psoriasis patients across all disease severities utilize topical drug products as their sole treatment or as an adjunct to their systemic treatment. However, 75 % of all patients treated with a topical agent fail to achieve significant, long-lasting improvement, and as a result, 40 % of diagnosed patients choose to forgo treatment. A 2013 survey analyzing the patient treatment landscape highlighted under treatment and non-treatment as the most significant issues in psoriasis; widespread patient dissatisfaction with current treatment options was cited as the primary driver.
[000193] SNA-125 represents one approach to the treatment (e.g., topical) of atopic dermatitis, psoriasis vulgaris, or both, as well as the substantial and undertreated condition of associated pruritus. Two positive controls will also be tested in this study: SNA-120; and DAIVONEX® Ointment 50 pg (calcipotriol)— a topical treatment for psoriasis vulgaris.
[000194] In this example SNA- 125 has been formulated for topical administration as an aqueous gel containing dimethyl sulfoxide (DMSO), propylene glycol, hydroxyethyl cellulose, benzyl alcohol, and trolamine at 2% w/w and 0.2% w/w strengths. A matching placebo vehicle with the same quantitative/qualitative composition with the exclusion of the active component has also been developed. Of note, the placebo vehicle was found to possess mild irritation potential in skin of subjects with a history of atopic dermatitis in a l4-day cumulative irritation assay.
[000195] The following adverse reactions have been identified for the treatment with DAIVONEX® 50 pg/g calcipotriol ointment 0.005 % (18):
Diseases of the skin and the subcutaneous cell tissue: Frequent (> 1 % - < 10 %): pruritus, burning, stinging, skin dryness, erythema, skin irritation/rash (incl. erythematous, maculopapulous or pustulous irritations),
Occasionally (> 0.1 % - <1 %): eczema, contact dermatitis, worsening of psoriasis, and
Very rarely (< 0.01 %): temporary change of the skin pigmentation (hyperpigmentation, depigmentation), temporary photosensitivity, hypersensitivity (including urticaria, periorbital or facial edema, angioedema), perioral or facial dermatitis; and
Metabolism and nutritional disturbances:
Very rarely (< 0.01 %): hypercalcemia, hypercalciuria.
[000196] One objective of this trial is to evaluate antipsoriatic efficacy and safety of SNA- 125 in topical formulations compared to topically applied reference formulations and vehicle in subjects with psoriasis vulgaris.
[000197] This is a 2-center, randomized, vehicle- and comparator-controlled, double-blind trial. Treatments will be randomly assigned to the test fields. All subjects will receive all treatments, with intra-individual comparison of treatments. Fifteen (15) male or female subjects of non- childbearing potential aged 18 years or older with chronic psoriasis vulgaris will be randomized to produce at least 12 evaluable cases at the end of the trial.
[000198] A total of 5 test fields located on the torso or the extremities will be examined per subject. The test fields will be treated occlusively for a 12-day treatment period (no IMP application on Day 7 and 12).
[000199] Treatment will be topical application of approximately 100 pL per test field (1.1 cm2) of the respective IMP once daily during a l2-day treatment period (10 treatments, no application on Days 7 and 12).
[000200] Investigational medicinal products to be administered are as follows: IMP 1 : 2% w/w SNA-125, daily dosage: approx. 2 mg SNA-125, total dosage for the treatment period: approx. 20 mg SNA- 125;
IMP 2: 0.2% w/w SNA-125, daily dosage: approx. 0.2 mg SNA-125, total dosage for the treatment period: approx. 2 mg SNA-125;
IMP 1 and IMP 2, total combined dosage of SNA-125: daily dosage: approx. 2.2 mg SNA-125, total dosage for the treatment period: approx. 22 mg SNA-125; IMP 3: 0.5% w/w SNA-120 (comparator), daily dosage: approx. 0.5 mg SNA-120, total dosage for the treatment period: approx. 5 mg SNA- 120;
IMP 4 (vehicle): active ingredient-free vehicle to IMP 1 and IMP 2;
IMP 5 (marketed comparator): DAIVONEX® 50 pg/g ointment (calcipotriol 0.005%), daily dosage: approx. 0.005 mg calcipotriol, total dosage for the treatment period: approx. 0.05 mg calcipotriol.
[000201] Details of the IMPs are given in Table 14 and Table 15.
Table 14.
Figure imgf000046_0001
Table 15.
Figure imgf000046_0002
Figure imgf000047_0001
[000202] All subjects will receive the same treatments. There will be no subdivision into treatment groups. A subject’s test fields will be numbered with 1 to 5. Figure 4 shows representatives of 2 of a number of possible scenarios. The actual distribution of plaques and test fields may vary individually. Randomization of the IMPs 1 to 5 will be performed using a Williams design. For each subject, a treatment sequence will be randomly assigned.
[000203] Approximately 100 pL of each IMP will be applied to a total of 5 test fields using special test chambers (Finn chambers, 12 mm inside 0, 14 mm outside 0). This is the amount required to completely fill the test chamber. The IMPs will be applied in the Finn chamber and seated in holes punched in a hydrocolloid dressing. The hydrocolloid dressing will be fixed before on the skin with adhesive patches containing the same holes for the chambers like the hydrocolloid dressing.
[000204] The chambers themselves will be fixed in place with LEUKOSILK® or the like and will be removed before each new application. The distance between the chambers must be at least 1.5 cm to exclude interactions with neighboring fields. The fields will be treated occlusively for a treatment period of 12 days. On 10 of the trial days treatments will be performed (Days 1 to 6 and 8 to 11). Before each new application, remaining preparation residues will be removed by gently cleansing each test field with a separate soft tissue. The hydrocolloid dressing will be renewed on Days 4 and 8.
[000205] In a subgroup of at least 5 subjects, a total of 12 punch biopsies per subject will be taken under local anesthesia for hematoxylin and eosin stain (H&E), immunohistochemistry (IHC) and biomarker analyses. At Baseline, 4 biopsies will be obtained from lesional untreated and non-lesional skin (two 4-mm biopsies from each skin type). On Day 12 (EoT) 8 biopsies will be obtained from test fields treated with IMP 1 (SNA-125 high dose), IMP 2 (SNA-125 low dose), IMP 3 (SNA-120) and IMP 4 (vehicle control) (two 4-mm biopsies from each test field). All biopsies will be excised after photographic documentation, sonography and clinical assessments. All skin biopsies will be processed and analyzed using established methods and protocols.
[000206] Blood pressure and pulse rate will be measured at screening, baseline and Day 12 (EoT). A physical examination will be performed at screening, Day 1 and on Day 12.
[000207] The trial is aimed to detect a treatment effect as compared to DAIVONEX® 50 pg/g ointment (calcipotriol 0.005%) within the planned trial design. A former in-house trial revealed a psoriatic infiltrate thickness reduction on Day 12 of about 51 % of the baseline level with a coefficient of variance (CV) below 0.97 for the difference to vehicle in change from baseline.
Summary of Results
[000208] Study subject disposition is presented in Table 16.
Table 16. Subject Disposition Category
N (%)
Enrolled 15 (100)
Treated 15 (100)
Completed Study (Visit 14) 15 (100)
Discontinued 0 (0)
Discontinued due to AE 0 (0)
[000209] The amount and duration of study drug exposure is summarized in Table 17.
Table 17. Study Drug Exposure SNA-125 2%
SNA-125 2% N = 15
No. treatments 10
Duration of exposure (days) 12
Total amount of study drug applied (mg) 20
SNA-125 0.2% N = 15
No. treatments 10 Duration of exposure (days) 12
Total amount of study drug applied (mg) 2
SNA- 120 0.5% N = 15
No. treatments 10
Duration of exposure (days) 12
Total amount of study drug applied (mg) 5
Calcipotriol 0.005% N = 15
No. treatments 10
Duration of exposure (days) 12
Total amount of study drug applied (mg) 0.05
[000210] Analyses of histology and biomarkers were conducted on biopsy samples obtained from lesional (LS) and non-lesional skin (NLS) from a subset of 11 subjects.
[000211] A total of 12, 4 mm punch, biopsies were taken per subject. Four biopsies were taken at baseline from lesional and non-lesional skin. Eight biopsies were taken at Day 12 (EoT) from treated lesional skin including 2 from each test field for 2% and 0.2% SNA-125, gel vehicle, and 0.5% SNA-120.
[000212] H&E and immunohistochemistry (IHC) results from one subject are shown in panel display in Figure 5. In aggregate, these data demonstrate an improvement on epidermal thickness and staining for various biomarkers. Specifically, 0.2 % SNA-125 had a reduction in epidermal thickness and K16 expression. Also, 0.5 % SNA-120 showed a reduction in K16 staining.
[000213] Figure 6 shows the aggregate epidermal thickness results for treated lesional skin at Day 12. There was a statistically significant improvement in epidermal thickness for 0.2 % SNA-125 compared to Baseline (day 1) lesional skin.
[000214] The normalization and expression controls for lesional and non- lesional skin in this study indicated the expected molecular signatures of psoriasis based on historical experience. Aggregate microarray (MA) expression data are presented in Figure 7 for each test article showing global up- and down-regulation of genes.
[000215] Improvement in gene expression results was attained with 0.2 % SNA- 125, where an approximate 11 % mean improvement in gene expression was observed. This level of response is consistent with very early (i.e., one week) treatment responses seen with some biologies (e.g., etanercept and ustekinumab) in separate experiments prior to their full clinical effect.
[000216] Figure 8 shows the results of disease (i.e. psoriasis) transcriptome (i.e., discrete set of co-modulated genes) analysis, quantification of improvement of different disease pathways. The best improvements were seen with 0.2 % SNA-125 and 0.5 % SNA- 120.
[000217] Subjects with 50 % improvement with any test article for keratin 16 (K16) (MA gene) and K16 (RTPCR marker) were considered“responders” to treatment. There were 3 subjects out of 11 subjects with biopsies who met this definition. The responder analysis of global gene expression data is shown in Figure 9. Responders showed higher overall % improvement than non-responders for all test articles (with evident increased variability - SD).
[000218] Conclusions: this exploratory Phase 1/2, double-blind, within-subject vehicle-controlled study in 15 subjects, using a psoriasis microplaque model, measured an effect on local tolerability, and histology and biomarkers with two doses of SNA-125 prototype gel applied once daily for 10 days. In this model, SNA-125 was well-tolerated and showed no safety signals; there were no application site treatment emergent adverse events (TEAEs) in any of the test fields. Histological and biomarker analysis showed a statistically significant reduction with SNA-125 0.2 % in epidermal thickness from baseline (-17 %, p<0.05), as well as modulation of certain psoriasis-relevant biomarkers and gene expression profiles. Histological and biomarker analyses indicate there was a signal for a drug effect with 0.2 % SNA-125 that was seen when using pathway- or whole transcriptome-related approaches, an effect that was statistically different from vehicle.
[000219] Overall, these data support impact of SNA-125 (e.g, the 0.2 % dose) on the histological characteristics and molecular signature of lesional psoriasis skin.

Claims

WHAT IS CLAIMED IS:
1. A topical gel formulation for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure, the formulation comprising:
a polymer conjugate of an indolocarbazole compound;
a preservative/solvent;
a solvent;
a penetration enhancer;
a gelling agent;
a pH adjusting agent; and
purified water q.s..
2. The gel formulation of Claim 1, wherein the polymer conjugate of an indolocarbazole compound is SNA-125.
3. The gel formulation of Claim 2, wherein the SNA-125 is present at about 0.05% to about 25% (w/w) of the gel formulation.
4. The formulation of Claim 2, wherein the SNA- 125 is present at about 0.5% to about 10% (w/w) of the gel formulation.
5. The formulation of Claim 2, wherein the SNA- 125 is present at about 5% (w/w) of the gel formulation.
6. The formulation according to any one of Claims 1 to 5, wherein the preservative/solvent is selected from benzoic acid, sorbic acid, boric acid, methylparaben, ethylparaben, propylparaben, butylparaben, sodium benzoate, sodium propionate, potassium sorbate, chlorobutanol, benzyl alcohol and phenyl ethyl alcohol, phenol, chlorocresol, o-phenyl phenol, benzalkonium chloride, cetyl pyridinium chloride, imidurea, thimerisal, propyl gallate, butylated hydroxyanisole (BHA), butylated hydroxy toluene (BHT), citric acid or disodium edetate.
7. The formulation according to any one of Claims 1 to 6, wherein the preservative/solvent is benzyl alcohol.
8. The formulation according to any one of Claims 1 to 7, wherein the preservative/solvent is present at about 0.1% to about 10% (w/w) of the gel formulation.
9. The formulation according to any one of Claims 1 to 8, wherein the preservative/solvent is present at about 1% (w/w) of the gel formulation.
10. The formulation according to any one of Claims 1 to 9, wherein the solvent is selected from ethanol, propylene glycol, glycerin or polyethylene glycol.
11. The formulation according to any one of Claims 1 to 10, wherein the solvent is propylene glycol.
12. The formulation according to any one of Claims 1 to 11, wherein the solvent is present at about 1% to about 30% (w/w) of the gel formulation.
13. The formulation according to any one of Claims 1 to 12, wherein the solvent is present at about 20% (w/w) of the gel formulation.
14. The formulation according to any one of Claims 1 to 13, wherein the penetration enhancer is DMSO.
15. The formulation according to any one of Claims 1 to 14, wherein the penetration enhancer is present at about 1% to about 30% (w/w) of the gel formulation.
16. The formulation according to any one of Claims 1 to 15, wherein the penetration enhancer is present at about 20% (w/w) of the gel formulation.
17. The formulation according to any one of Claims 1 to 16, wherein the gelling agent is HEC.
18. The formulation according to any one of Claims 1 to 17, wherein the gelling agent is present at about 0.5% to about 5% (w/w) of the gel formulation.
19. The formulation according to any one of Claims 1 to 18, wherein the gelling agent is present at about 1.25% (w/w) of the gel formulation.
20. The formulation according to any one of Claims 1 to 19, wherein the pH adjusting agent is trolamine.
21. The formulation according to any one of Claims 1 to 20, wherein the pH adjusting agent is added q.s. to bring the final gel formulation to a pH of greater than or equal to about 5.0.
22. A method of treating a skin condition associated with kinase signaling, the method comprising:
applying, or instructing application of, a topical formulation to a skin region, wherein said formulation inhibits kinase signaling in said skin region, and thereby treats the skin condition;
wherein said topical formulation is the gel formulation of Claim 1.
23. The method of Claim 22, wherein the polymer conjugate of an indolocarbazole is SNA- 125.
24. The method of Claim 22 or 23, wherein the kinase is TrkA, a JAK kinase selected from JAK1, JAK2 or JAK3, a tyrosine kinase 2 (TYK2), or a mitogen- activated protein kinase kinase selected from MAP2K or MAP2K3.
25. The method according to any one of Claims 22 or 24, wherein the skin condition is atopic dermatitis, psoriasis, pruritus or any other dermatologic condition.
PCT/US2018/066403 2017-12-20 2018-12-19 Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure WO2019126270A1 (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201762608366P 2017-12-20 2017-12-20
US62/608,366 2017-12-20
US201862723347P 2018-08-27 2018-08-27
US62/723,347 2018-08-27
US201862732538P 2018-09-17 2018-09-17
US62/732,538 2018-09-17
US201862774729P 2018-12-03 2018-12-03
US62/774,729 2018-12-03

Publications (1)

Publication Number Publication Date
WO2019126270A1 true WO2019126270A1 (en) 2019-06-27

Family

ID=66992794

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/066403 WO2019126270A1 (en) 2017-12-20 2018-12-19 Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure

Country Status (1)

Country Link
WO (1) WO2019126270A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022067042A1 (en) * 2020-09-24 2022-03-31 Regents Of The University Of Minnesota Cryoprotective compositions and methods for protection of a surgical site during cryosurgery

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080193517A1 (en) * 2005-08-25 2008-08-14 Creabilis Therapeutics S.P.A. Polymer Conjugates of K-252A and Derivatives Thereof
US20110112121A1 (en) * 2009-07-06 2011-05-12 Joerg Berghausen Pharmaceutical Compositions and Solid Forms
US20150125474A1 (en) * 2013-10-11 2015-05-07 Asana Biosciences, Llc Protein-polymer-drug conjugates

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080193517A1 (en) * 2005-08-25 2008-08-14 Creabilis Therapeutics S.P.A. Polymer Conjugates of K-252A and Derivatives Thereof
US20110112121A1 (en) * 2009-07-06 2011-05-12 Joerg Berghausen Pharmaceutical Compositions and Solid Forms
US20150125474A1 (en) * 2013-10-11 2015-05-07 Asana Biosciences, Llc Protein-polymer-drug conjugates

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HEINEN ET AL.: "Trk inhibition reduces cell proliferation and potentiates the effects of chemotherapeutic agents in Ewing sarcoma", ONCOTARGET, vol. 7, no. 23, 26 April 2016 (2016-04-26), XP055615967 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022067042A1 (en) * 2020-09-24 2022-03-31 Regents Of The University Of Minnesota Cryoprotective compositions and methods for protection of a surgical site during cryosurgery

Similar Documents

Publication Publication Date Title
Yokota et al. Influence of nanoparticle size on the skin penetration, skin retention and anti-inflammatory activity of non-steroidal anti-inflammatory drugs
Chen et al. Topical delivery of Cyclosporine A into the skin using SPACE-peptide
US11607399B2 (en) Pharmaceutical formulation for histone deacetylase inhibitors
US20080311223A1 (en) Injectable polymer-lipid blend for localized drug delivery
JP2001512487A (en) Anti-cell proliferating agent gel delivery vehicle
JP2009504751A (en) Topical preparation of histone deacetylase inhibitor and method of use thereof
US20220096459A1 (en) Resiquimod topical and injectable compositions for the treatment of neoplastic skin conditions
US9872912B2 (en) Method for treating pancreatic cancer
Jiang et al. Self-assembled nanoparticles with bilirubin/JPH203 alleviate imiquimod-induced psoriasis by reducing oxidative stress and suppressing Th17 expansion
Gorantla et al. Hyaluronic acid-coated proglycosomes for topical delivery of tofacitinib in rheumatoid arthritis condition: Formulation design, in vitro, ex vivo characterization, and in vivo efficacy studies
Xia et al. Transdermal administration of ibuprofen-loaded gel: preparation, pharmacokinetic profile, and tissue distribution
Jung et al. H2O2-activatable hybrid prodrug nanoassemblies as a pure nanodrug for hepatic ischemia/reperfusion injury
WO2020023389A1 (en) Reduced exposure compositions modulating therapeutic targets
WO2019126270A1 (en) Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure
EP3187172A1 (en) Cyclosporine a topical compositions
EP4013418A1 (en) Uracil dermal pharmaceutical formulation
Doppalapudi et al. Fenoldopam mesylate for treating psoriasis: a new indication for an old drug
CN109069472A (en) Medical composition containing producing high-molecular drug
WO2019143971A2 (en) Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure
WO2020023387A1 (en) Uses of polymer conjugates of indolocarbazole compounds with reduced exposure
US20190209472A1 (en) Formulations for dermal delivery of polymer conjugates of indolocarbazole compounds with reduced exposure
Nayak et al. Formulation, optimization and evaluation of ibuprofen loaded menthosomes for transdermal delivery
WO2014076642A1 (en) Topical composition for the transepidermal or transdermal delivery of paracetamol
Ferreira Nanostructured lipid carriers: a new approach for psoriasis dermal therapy
EP3621657A1 (en) Uses of polymer conjugates of indolocarbazole compounds with reduced exposure

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: 18892892

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18892892

Country of ref document: EP

Kind code of ref document: A1