WO2014015107A1 - Dosing regimen for janus kinase (jak) inhibitors - Google Patents

Dosing regimen for janus kinase (jak) inhibitors Download PDF

Info

Publication number
WO2014015107A1
WO2014015107A1 PCT/US2013/051015 US2013051015W WO2014015107A1 WO 2014015107 A1 WO2014015107 A1 WO 2014015107A1 US 2013051015 W US2013051015 W US 2013051015W WO 2014015107 A1 WO2014015107 A1 WO 2014015107A1
Authority
WO
WIPO (PCT)
Prior art keywords
therapeutically effective
mammal
effective dose
jak
day
Prior art date
Application number
PCT/US2013/051015
Other languages
French (fr)
Inventor
Andrea J. GONZALES
Sallie B. COSGROVE
Phyllis B. MALPAS
Michael Rolf STEGEMANN
Wendy Turner COLLARD
Original Assignee
Zoetis Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48901189&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014015107(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to RS20190051A priority Critical patent/RS58242B1/en
Priority to JP2015523243A priority patent/JP6022063B2/en
Priority to IN370DEN2015 priority patent/IN2015DN00370A/en
Priority to NZ703152A priority patent/NZ703152A/en
Priority to MX2015000871A priority patent/MX360857B/en
Priority to AU2013292547A priority patent/AU2013292547B2/en
Priority to KR1020157001085A priority patent/KR20150028299A/en
Priority to EP13742573.2A priority patent/EP2874630B1/en
Priority to CA2878867A priority patent/CA2878867C/en
Priority to SI201331328T priority patent/SI2874630T1/en
Priority to ES13742573T priority patent/ES2707627T3/en
Priority to LTEP13742573.2T priority patent/LT2874630T/en
Priority to PL13742573T priority patent/PL2874630T3/en
Priority to CN201380038563.3A priority patent/CN104470525A/en
Priority to BR112015000808A priority patent/BR112015000808A2/en
Priority to KR1020177007913A priority patent/KR20170034949A/en
Priority to US14/415,047 priority patent/US9522151B2/en
Priority to DK13742573.2T priority patent/DK2874630T3/en
Application filed by Zoetis Llc filed Critical Zoetis Llc
Publication of WO2014015107A1 publication Critical patent/WO2014015107A1/en
Priority to ZA2015/00134A priority patent/ZA201500134B/en
Priority to HK15109423.1A priority patent/HK1209314A1/en
Priority to HRP20182088TT priority patent/HRP20182088T1/en
Priority to CY20191100203T priority patent/CY1121436T1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dermatology (AREA)
  • Immunology (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

The present invention provides a method for treating allergic dermatitis, atopic dermatitis, or one or more symptoms thereof in a mammal in need, which method comprises administering to the mammal a first therapeutically effective dose of a Janus Kinase (JAK) inhibitor twice a day for a number of days sufficient to ease or eliminate one or more clinical signs in the mammal, followed by a second therapeutically effective dose of the JAK inhibitor at a reduced frequency.

Description

DOSING REGIMEN FOR JANUS KINASE (JAK) INHIBITORS
FIELD OF THE INVENTION
This invention relates to Janus Kinase (JAK) inhibitors. This invention also relates to diseases and conditions such as itch, pruritus, and dermatitis. This invention also relates to the administration and dosing of certain compounds having activity as JAK inhibitors.
BACKGROUND OF THE INVENTION
Protein kinases are families of enzymes that catalyze the phosphorylation of specific residues in proteins, broadly classified into tyrosine and serine/threonine kinases. Inappropriate kinase activity, arising from mutation, over-expression, or inappropriate regulation, dys-regulation or de-regulation, as well as over- or underproduction of growth factors or cytokines has been implicated in many diseases, including but not limited to cancer, cardiovascular diseases, allergies, asthma and other respiratory diseases, autoimmune diseases, inflammatory diseases, bone diseases, metabolic disorders, and neurological and neurodegenerative disorders such as Alzheimer's disease. Inappropriate kinase activity triggers a variety of biological cellular responses relating to cell growth, cell differentiation, survival, apoptosis, mitogenesis, cell cycle control, and cell mobility implicated in the aforementioned and related diseases.
Thus, protein kinases have emerged as an important class of enzymes as targets for therapeutic intervention. In particular, the JAK family of cellular protein tyrosine kinases (JAK-1 , JAK-2, JAK-3, and Tyk-2) play a central role in cytokine signaling (Kisseleva et al, Gene, 2002, 285, 1 ; Yamaoka et al. Genome Biology 2004, 5, 253)). Upon binding to their receptors, cytokines activate JAK which then phosphorylate the cytokine receptor, thereby creating docking sites for signaling molecules, notably, members of the signal transducer and activator of transcription (STAT) family that ultimately lead to gene expression. Numerous cytokines are known to activate the JAK family.
SUMMARY OF THE INVENTION
The present invention provides a method for treating allergic dermatitis, atopic dermatitis, or one or more symptoms thereof in an animal, particularly a mammal in need, which method comprises administering to the mammal a first therapeutically effective dose of a Janus Kinase (JAK) inhibitor twice a day for a number of days sufficient to ease or eliminate one or more clinical signs in the mammal, followed by a second therapeutically effective dose of the JAK inhibitor at a reduced frequency.
In one embodiment, the first therapeutically effective dose and the second therapeutically effective dose are administered orally. In another embodiment, the first therapeutically effective dose is administered parenterally and the second therapeutically effective dose is administered orally.
The invention also provides a method for treating allergic dermatitis, atopic dermatitis, or one or more symptoms thereof in a mammal in need as described herein, ompound of formula I:
Figure imgf000004_0001
I
or a pharmaceutically acceptable salt thereof, wherein 1 is Ci_4 alkyl optionally substituted with hydroxyl. In one embodiment, R1 is methyl. In another embodiment, R1 is ethyl or cyclobutyl.
In another embodiment, the JAK inhibitor is
Figure imgf000004_0002
pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methanesulfonamide, or a pharmaceutically acceptable salt thereof.
The invention also provides a method for treating atopic dermatitis or pruritus in a mammal in need comprising administering to the mammal a first therapeutically effective dose of /V-methyl-1 -{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide, or a pharmaceutically acceptable salt thereof, orally, twice a day for a number of days sufficient to ease or eliminate one or more clinical signs in the mammal, followed by a second therapeutically effective dose, orally, of /V-methyl-1 -{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide, or the pharmaceutically acceptable salt thereof at a reduced frequency.
In one embodiment of the invention, the mammal is a companion animal selected from a dog and a cat. In another embodiment, the mammal is a dog. In one embodiment of the method of the invention for treating atopic dermatitis or pruritus in a mammal by administering a JAK inhibitor, for example
Figure imgf000005_0001
-{trans-4- [methyl(7H-pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methanesulfonamide, or a pharmaceutically acceptable salt thereof, the first therapeutically effective dose is from about 0.4 to about 0.6 mg/kg body weight of the mammal and is administered twice a day. In this embodiment, the second therapeutically effective dose is preferably from about 0.4 to about 0.6 mg/kg body weight of the mammal and is administered at the reduced frequency. In a further embodiment, the number of days of administration of the first therapeutically effective dose is from 1 day to 42 days, preferably 14 days.
This invention also provides method for treating a disease or condition caused by or associated with an immune system dysfunction or immune system dysregulation in a mammal in need, which method comprises orally administering to the mammal a therapeutically effective amount of a compound of formula I:
Figure imgf000005_0002
I
or a pharmaceutically acceptable salt thereof, wherein 1 is Ci_4 alkyl optionally substituted with hydroxyl, two times per day for a period of from 1 day to 42 days (6 weeks), followed by administering to the mammal the therapeutically effective amount one time per day. Preferably, the compound is /V-methyl-1 -{frans-4-[methyl(7H- pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methanesulfonamide. Preferably, in this method, the therapeutically effective amount of the compound of formula I is administered to the mammal two times per day for a period of from 1 to 14 days, followed by administering to the mammal the therapeutically effective amount one time per day. In a further embodiment, the disease or condition is an allergic reaction or eczema.
The invention also provides for the method for treating a disease or condition caused by or associated with an immune system dysfunction or immune system dysregulation in a mammal in need as recited above, wherein the therapeutically effective amount of the compound of formula I is from about 0.4 mg/kg body weight of the mammal to about 3.0 mg/kg body weight of the mammal. The dose may be from 0.1 to 2 mg/kg, or 0.2 to 1 mg/kg, or 0.3 to 0.8 mg/kg. Preferably, the therapeutically effective amount of the compound of formula I is from about 0.4 mg/kg body weight of the mammal to about 0.6 mg/kg body weight of the mammal.
The invention also provides a method for improving the therapeutic ratio of a Janus Kinase-1 (JAK-1 ) inhibitor, comprising: administering to a mammal, over a period of at least 5-days, a plurality of therapeutically effective doses of said JAK-1 inhibitor sufficient to inhibit interleukins dependent on JAK-1 ; wherein the plurality of therapeutically effective doses do not reach peak drug levels of the JAK-1 inhibitor above the IC50 for a hematopoietic cytokine. The invention further provides a method for maintaining the inhibition corridor in a mammal comprising administering the compound(s) described herein according to the dosing regimen described herein.
In a more particular embodiment, the interleukins are selected from the group consisting of IL-31 , IL-4, IL-2, IL-6 and IL-13. In a more particular embodiment, the hematopoietic cytokines are selected from the group consisting of erythropoietin (EPO) or granulocyte colony-stimulating factor (GM-CSF). In another embodiment, the period of administration, is at least 10 days, 12 days, or preferably 14 days. BRIEF DESCRIPTION OF THE FIGURES
Figure 1. Drug concentrations of Compound 1 and relationship to inhibition of cytokine function.
Figure 2. Least-squares mean plasma concentration time profiles of Compound 1 in beagle dogs following oral twice daily administration (Day 0, 21 ) and once daily administration (Day 53, 168) of 0.6 mg/kg, 1.8 mg/kg and 3.0 mg/kg.
DETAILED DESCRIPTION
"Mammal" refers to humans or non-human animals, including livestock and companion animals. The phrase "companion animal" refers to an animal kept as a pet. Examples of companion animals include cats, dogs, and horses. The term "livestock" refers to animals reared or raised in an agricultural setting to make products such as food or fiber, or for its labor. In some embodiments, livestock are suitable for consumption by mammals, for example humans. Examples of livestock animals include cattle, goats, horses, pigs, sheep, including lambs, and rabbits. Also included within the definition of "mammal", for purposes of this invention, are birds, such as chickens, ducks and turkeys.
"Therapeutic ratio" as used herein refers to a comparison of the therapeutic effect to the toxicity or adverse effect of the drug. A therapeutic effect can be attained by reduction in the disease state or other forms of "treatment" as defined below. It can also be achieved by modulation of a particular target, such as inhibition of interleukins implicated in a disease state, such as atopic dermatitis. Accordingly, improving therapeutic ratio can occur by, for example, modifying the dosing regimen such that inhibition of interleukins associated with a target disease state occurs, while modulation of other cytokines associated with toxicity, such as EPO, GM-CSF, IL-12, IFN-alpha, or IL-23 is minimized, referred to herein as the "inhibition corridor". By carefully maintaining the inhibition corridor between toxic cytokines and efficacy-related cytokines (as depicted in Figure 1 ), positive drug effects are maximized while minimizing or eliminating side-effect (i.e. improving the therapeutic ratio).
"Treating" or "treatment" as used herein means controlling, treating, or preventing the progression of the indicated condition or disease. The term "controlling", "treating" or "treatment" of a condition or disease includes: (1 ) preventing the condition or disease, i.e. causing the clinical symptoms or signs of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms/signs of the disease; (2) inhibiting the disease, i.e., arresting or reducing the progression of the disease or its clinical symptoms or signs; or (3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms or signs
The JAK inhibitor is preferably a compound as described in US 2002/0019526 (Publication Date February 14, 2002, from United States Patent Application 09/956,645, filed on September 19, 2001 ), the teachings and contents of which are incorporated herein in their entirety by reference.
JAK inhibitor is a compound of formula I:
Figure imgf000007_0001
I
or a pharmaceutically acceptable salt thereof, wherein 1 is Ci_4 alkyl optionally substituted with hydroxy. Compounds of formula I, their synthesis, and their use as JAK inhibitors are described in US 2010/0075996 A1 (Publication Date March 25, 2010, from United States Patent Application 12/542,451 , filed August 17, 2009), the teachings and contents of which are hereby incorporated by reference in their entirety.
The "pharmaceutically acceptable salt" can be any salt suitable for pharmaceutical use in a mammal, preferably the acetate, ascorbate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, etoglutarate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, glycerophosphate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate or trifluoroacetate salt. Preferably the pharmaceutically acceptable salt is the maleate (or maleic acid) salt.
Preferably the compound of formula I is /V-methyl-1-{frans-4-[methyl(7H- pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methanesulfonamide or a pharmaceutically acceptable salt thereof. In a preferred embodiment, the compound of formula I is /V-methyl-1-{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide (referred to herein as compound 1 ), preferably the maleic acid salt
The phrases "therapeutically effective amount" and "therapeutically effective dose" in general, unless otherwise specified, mean an amount of a compound that, when administered to a mammal for treating a condition or disease as recited, is sufficient to effect treatment of the condition or disease. More specifically, a therapeutically effective amount or dose means an amount of compound that, when administered according to a regimen as recited, is effective to prevent, alleviate or ameliorate symptoms or signs of a disease or condition or prolong the survival of the subject being treated. The "therapeutically effective amount" or "dose" may vary depending on the compound, the disease and its severity, and depending on the age, weight, and other such factors of the mammal to be treated. Generally, a therapeutically effective amount of a JAK inhibitor for purposes of the present invention is from about 0.01 to about 100 mg/kg of body weight per day, preferably about 0.1 to about 10 mg/kg of body weight per day.
As described herein, the method for treating a disease or condition caused by or associated with an immune system dysfunction or dysregulation involves a first administration phase of a higher daily therapeutic dose to the mammal, followed by a second administration phase wherein the daily therapeutically effective dose of the JAK inhibitor is lower than the dose in the first phase. The daily therapeutically effective dose during the second administration phase can be at a reduced frequency relative to the dose during the first administration phase.
Preferably the "first therapeutically effective dose", which is the dose given during the first administration phase, is administered in divided doses, e.g. two times per day. Preferably the first therapeutically effective dose is, for example, from about 0.05 to about 3 mg/kg two times per day (BID), for a total daily dose of from about 1 mg/kg to about 6 mg/kg per day. More preferably, the first therapeutically effective dose is from about 0.1 to about 1 mg/kg BID, and even more preferably from about 0.4 to about 0.6 mg/kg BID. In another embodiment, the first therapeutically effective dose is from about 0.4 to about 3 mg/kg BID. In another embodiment, the first therapeutically effective dose is about 0.6 mg/kg, 1.8 mg/kg, or 3/0 mg/kg BID. In another embodiment, the first therapeutically effective dose is from about 0.2-0.3 mg/kg BID.
In one embodiment, the second therapeutic dose is the same as the first therapeutic dose, except that it is given at reduced frequency, for example one time per day (SID), relative to the frequency of the first therapeutic dose. In another embodiment, the second therapeutic dose is from about 0.05 to about 3 mg/kg one time per day (SID). More preferably, the second therapeutically effective dose is from about 0.1 to about 1 mg/kg SID, and even more preferably from about 0.4 to about 0.6 mg/kg SID. In another embodiment, the second therapeutically effective dose is from about 0.4 to about 3 mg/kg SID. In another embodiment, the second therapeutically effective dose is about 0.6 mg/kg, 1.8 mg/kg, or 3/0 mg/kg SID. In another embodiment, the second therapeutically effective dose is from about 0.2-0.3 mg/kg SID.
According to the method of the invention, the first therapeutically effective dose is given for a period of time, for example a number of days, sufficient to ease or eliminate one or more clinical signs of the disease or condition, for example the allergic dermatitis or atopic dermatitis. This can be conveniently referred to as a "first administration phase". The period of time sufficient to ease or eliminate one or more clinical signs of the disease or condition can be determined base on observation of reduction in the clinical sign or signs, for example using recognized criterion, as described in further detail herein. Thereafter, the period of time for the first administration phase can be set. In one embodiment, the first administration phase is from about 3 months. In another embodiment the first administration phase is 6 weeks, 4 weeks or 3 weeks. In another embodiment, the first administration phase is about 14 days. In another embodiment, the first administration phase is 10 days, 7 days, or 1 , 2, 3, 4, 5, or 6 days.
Following the first administration phase, in the methods of the subject invention for treating a disorder or condition caused by, or associated with an immune system dysfunction or dysregulation, for example, atopic dermatitis or allergic dermatitis, a second therapeutically effective dose of the JAK inhibitor is administered to the mammal, and the second therapeutically effective dose is 1 ) a reduced daily dose relative to the daily dose of the first administration phase, and/or 2) a reduced frequency relative to the frequency of administration of the first therapeutically effective dose. As described, in one embodiment, the first therapeutic dose is BID and the second therapeutic dose is SID.
The route of administration for the first administration phase can be different from the route of administration for the second administration phase. For example, the route of administration for the first administration phase may be parenteral, and the route of administration for the second phase may be oral.
The term "clinical sign" as used herein refers to an observable or measureable condition or behavior in the mammal that is indicative of the disease, condition or symptom. Clinical signs may be those symptoms, conditions, or behaviors that are measured in known or established diagnostic assessments. For example, diagnostic assessments for a determination of allergic dermatitis or atopic dermatitis can be made by a Visual Analog Scale (VAS) Score or a clinical assessment of condition, or by an established scoring system such as the Canine Atopic Dermatitis Extent and Severity Index (CADESI) Score. Non-limiting examples of some clinical signs for atopic dermatitis and allergic dermatitis, that may be used sometimes in such assessments or scoring systems, include: itching, ranging from extremely severe (as demonstrated, in the case of a companion animal such as a dog, by scratching, chewing, licking almost continuously, regardless of what else is happening), to severe (as demonstrated by prolonged episodes of itching while awake, and itching at night and/or while eating, playing or exercising), to moderate (as demonstrated by frequent episodes of itching), to very mild (occasional episodes of itching); presence of pustules or epidermal collarets; presence of skin lesions; pruritus; erythema; erosions, excoriations and/or self-induced alopecia; presence of papules and/or crusts; lichenification and/or hyperpigmentation.
A "symptom" of a disease or condition is any of those symptoms known by a person of ordinary skill in the art as being associated with the disease or condition. In the case of atopic dermatitis, allergic dermatitis, flea allergy dermatitis, and sarcoptic mange, symptoms include, for example: pruritus, itch, and skin lesions.
In many cases, a "symptom" of a disease or condition, such as atopic dermatitis or allergic dermatitis is also a "clinical sign".
In the case of allergic dermatitis, the allergic dermatitis may be flea allergy dermatitis, i.e. "FAD" (also called "flea allergic dermatitis", "flea bite dermatitis" ("FBD"), or "flea-associated dermatitis), food allergy dermatitis, contact dermatitis, or allergic dermatitis associated with Sarcoptes scabiei (i.e. sarcoptic mange).
Other indications and conditions that can be treated by the methods including the dosing regimens described herein include any indications or conditions treatable by administration of a JAK inhibitor, including those involving Janus Kinase-1 , Janus Kinase-2 or Janus Kinase-3. Such indications and conditions include organ transplant, lupus, multiple sclerosis, rheumatoid arthritis, psoriasis, Type I diabetes and complications from diabetes, cancer, asthma, atopic dermatitis, autoimmune thyroid disorders, ulcerative colitis, Crohn's disease, Alzheimer's disease, leukemia, osteoarthritis, control of pruritus, chronic respiratory disease and other indications where immunosuppression or immunomodulation would be desirable.
The "administration" of the JAK inhibitor according to the methods described herein can be administration orally, parenterally, topically, rectally, transmucosally, or intestinally. Parenteral administrations include indirect injections to generate a systemic effect or direct injections to the afflicted area. Topical administrations include the treatment of skin or organs readily accessible by local application, for example, eyes or ears. It also includes transdermal delivery to generate a systemic effect. The rectal administration includes the form of suppositories. The preferred routes of administration are oral and parenteral, with oral being most preferred.
In the methods described herein, the JAK inhibitor can be administered in dosage forms corresponding to the selected route of administration. The pharmaceutical compositions of the JAK inhibitors can be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compound into preparations, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Pharmaceutically acceptable excipients and carriers are generally known to those skilled in the art and are thus included in the instant invention. Such excipients and carriers are described, for example, in "Remingtons Pharmaceutical Sciences" Mack Pub. Co., New Jersey (1991 ). The dosage form can be, for example, for oral administration: tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents, fillers, lubricants or wetting agents; liquid preparations for oral administration such as solutions, syrups or suspension prepared by conventional means with conventional pharmaceutical excipients.. For buccal administration, the dosage form may take the form of tablets or lozenges. The tablets may be chewable and/or flavored. For parenteral administration, the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles; or the JAK inhibitor may be in powder form for reconstitution or in concentrated liquid form for subsequent dilution before administration. Rectal dosage forms may be conventional suppositories or retention enemas. Topical forms may be ointments, salves or transdermal patches. The JAK inhibitor may also be in a dosage form suitable for intranasal administration, or administration by inhalation.
The following Examples illustrate the methods and dosing regimens of the invention, but they are not to be construed as limiting the invention as fully described in the specification and recited in the claims.
EXAMPLES
Pruritus Studies
A study was conducted in client owned dogs. Fifty-six dogs, greater than 1 year of age, weighing 2-50 kg, with a history of chronic atopic dermatitis were enrolled at three veterinary schools. The study design is illustrated in the following Table 1 :
Table 1
Figure imgf000013_0001
All site personnel were masked to treatment group assignment. Placebo capsules were identical in appearance to compound capsules
2 Within each clinic, animals were blocked on order of enrollment with a block consisting of two animals
3 +1 day
4 Telephone call
5/V-methyl-1 -{frans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate
The reduction in owner assessed VAS pruritus scores were significantly ( p≤ 0.07) different for the compound-treated dogs compared with the placebo treated dogs at Day 1 , 14 and 28. The reduction in investigator assessed CADESI-02 skin lesion scores were significantly (p = 0.272) different for the compound-treated dogs compared to the placebo-treated.
In the following pruritus studies the owners had given consent and had completed a survey indicating that their dog exhibited moderate to severe itching. Investigators attributed pruritus to one or more of the following: allergic dermatitis, atopic dermatitis, food allergy, contact allergy, flea allergy, sarcoptic mange. Dogs were six months of age or older, weighed a minimum of 3 kg and were physically healthy apart from their pruritic condition. Dogs were flea-free, and appropriate preventatives and treatments were used throughout the studies.
The studies were designed as follows in Table 2 and Table 3.
Table 2 (Pruritus Study A)
Figure imgf000014_0001
capsules were identical in appearance to compound capsules
2 Within each clinic, animals were blocked on order of enrollment with a block consisting of two animals
3 +3 day
4 Treatment and Study Visits could continue through Day 28 ± 2 if underlying condition had not resolved
5/V-methyl-1-{frans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate
Table 3 (Pruritus Study B)
Figure imgf000014_0002
The Owner and Veterinarian were masked to treatment group assignment. The Dispenser (technician) was not masked to treatment group assignment
2 Within each clinic, animals were blocked on order of enrollment with a block consisting of two animals
3 +1 day
4 Treatment and Study Visits could be discontinued after Day 7 ± 1 if underlying condition had resolved 5N-methyl-1-{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate
In the Pruritus Study A, the owner VAS scores by day were lower each day starting on day 1 for T02 compared to T01. By day 7, the LS mean VAS score was about 25 mm for T02, whereas for T01 (placebo), the LS mean VAS score was about 55 mm. Note, at time zero, the LS mean VAS score for both T01 and T02 was about 75 mm. In the Pruritus Study B, the owner VAS scores over the 14 day period decreased for both T01 (prednisolone) and T02 (compound 1 , i.e. /V-methyl-1-{fra/is-4-[methyl(7H- pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methanesulfonamide maleate). At time point zero, both T01 and T02 had LS mean VAS scores of about 75 mm. At day 14, T01 has an LS mean VAS score of about 10 mm, and T02 has an LS mean VAS score of about 18 mm.
Atopic Dermatitis
In the following atopic dermatitis field studies, the owners had given consent and had completed a survey indicating that their dog exhibited either moderate to severe or mild to severe itching or dermatitis. Minimum CADESI scores were assigned by a dermatologist or veterinarian (CADESI-01 score of 25 or CADESI -3 score of 60). Dogs were either 1 year of age or 6 months of age or older and weighed a minimum of 3 kg and were physically healthy apart from their atopic disease. The dogs had at least a 1 year or 6 month documented history of chronic non-seasonal atopic dermatitis. The dogs were flea free and appropriate preventatives and treatments were used throughout the study.
Atopic Dermatitis "High Dose" Studies
In these studies, animals were administered from 0.4-0.6 mg/kg body weight of a JAK inhibitor BID for up to 1 12 days. Table 4
Figure imgf000016_0001
caplets were identical in appearance to compound caplets
2 Within each clinic, animals were blocked on order of enrollment with a block consisting of four animals
3 ±2-3 days
4 Dogs failing to show clinical improvement permitted to move into open-label (un-masked ) study
5/V-methyl-1-{frans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate
Table 5
Figure imgf000016_0002
1The Owner and Veterinarian were masked to treatment group assignment. The Dispenser (technician) was not masked to treatment group assignment
2 Within each clinic, animals were blocked on order of enrollment with a block consisting of two animals
3 ±2-7 days (depending on Study Visit) 4 Treatment and Study Visits could be discontinued at any time
5/V-methyl-1-{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate
A dose regimen of 0.4-0.6 mg/kg BID showed excellent efficacy for the control of atopic dermatitis including pruritus and was safe for up to 90 to 1 12 days of treatment in client-owned dogs. This same regimen was safe for up to 90 days at elevated dosages. However, for long term administration, the BID dosing regimen could not be supported with an adequate margin of safety. Therefore, a dose selection study was conducted, as described below, to evaluate alternative dosing regimens.
A high dose (3 mg/kg BID) was given to 6 month old laboratory dogs. The dogs showed clinical signs of integument (7/8 dogs with demodex). At week 14, male dog showed pneumonia, peritonitis, pleuritis consistent with bacterial infection; lymphoid depletion; lymphadenitis; mild inflammation of choroid plexus and hepatitis. Female dog, at week 14, showed fever, demodex, pyoderma, and some pneumonia.
Although the 0.6 mg/kg BID group showed few effects, the bacterial and parasitic infection in the 3 mg/kg BID dose group did not support chronic use BID.
Atopic Dermatitis Dose Selection Study
Figure imgf000017_0001
Table 6 (Atopic Dermatitis Dose Selection Study)
1 All site personnel were masked to treatment group assignment. Placebo caplets were identical in appearance to compound 1 caplets
2 Within each clinic, animals were blocked on order of enrollment with a block consisting of four animals
3 ±2 days
4 Dogs failing to show clinical improvement permitted to move into open-label (unmasked ) study
5/V-methyl-1-{frans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate
6 With placebo administered SID for first 14 days to ensure masking
Owner VAS scores for atopic dermatitis over the 1 12 days of study were in the following order from highest VAS score (most atopic dermatitis) to lowest VAS score: T01. T04, T03, and T02. Investigator CADESI score over the 1 12 days of study were in the following order from highest CADESI score (highest demonstration of atopic dermatitis) to lowest CADESI score: T01 (placebo), T04 (0.2-0.3 mg/kg SID compound 1 ), T03 (0.4-0.6 mg/kg SID compound 1 ), and T02 (0.4-0.6 mg/kg BID for 14 days followed by 0.4-0.6 mg/kg SID thereafter).
Target Animal Safety Studies
A target animal safety program (8 studies) was conducted. 131 laboratory-bred dogs were exposed to /V-methyl-1-{frans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate (i.e. N-methyl-1-{frans-4-[methyl(7/- - pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methanesulfonamide maleic acid salt maleic acid salt). Doses ranged from 0.5 mg/kg/day (0.25 mg/kg BID) to 18 mg/kg/day (9 mg/kg BID). Duration of exposure ranged from 10 days to 6 months. Recovery periods were incorporated into two studies. A series of early studies were designed to ensure the safety of client-owned dogs in the field safety and efficacy studies.
In a "margin of safety" study, the following treatments were given: Treatment Number of Dose (oral) Regimen Regimen Dosing
Animals1 Weeks 1 -6 Weeks 7-26 Days
(M/F)
T01 4/4 0.0 mg/kg 2 per day 1 per day 180
T02 4/4 0.6 mg/kg 2 per day 1 per day 180
T03 4/4 1.8 mg/kg 2 per day 1 per day 180
T04 4/4 3.0 mg/kg 2 per day 1 per day 180
Results of the "margin of safety" study were no observed deaths or other serious adverse events. Test article and dose related clinical signs were primarily seen grossly in the exacerbation of interdigital furunculosis with associated peripheral lymphadenopathy and the occasional development of papillomas. From this study, we concluded oral administration of /V-methyl-1-{f''ans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin- 4-yl)amino]cyclohexyl}methanesulfonamide in dogs, BID for 6 weeks followed by SID (one time per day) for 20 weeks at 0.6, 1.8, or 3.0 mg/kg for a total of 26 weeks (6 months) was well tolerated at all dose multiples. The test article effects in all groups were consistent with the pharmacological action of the drug class, and most effects were mild and non-progressive. Chronic use is supported in the population of dogs greater than 1 year old.
Results and Discussion
Any immune modulator may increase susceptibility to infections (dose dependent). Bacterial and fungal infections of the skin were the most common type of infection reported in dogs in the field safety and efficacy studies; these responded to appropriate antimicrobial therapy. In a high dose target animal safety study, in dogs less than one year old, at elevated doses, parasitic infestations (demodicosis) and pneumonia were observed. Demodicosis was reported in two dogs and pneumonia (attributable to a pulmonary mass) on one dog in the field safety and efficacy studies.
In summary, in the control or treatment of pruritus associated with allergic dermatitis and the control of atopic dermatitis in dogs, using a JAK inhibitor, /V-methyl- 1-{frans-4-[methyl(7/- -pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate, a dosing regimen of 0.4-.6 mg/kg BID for 14 days followed by SID thereafter is supported. The target animal safety studies support the long-term and chronic administration of /V-methyl-1-{frans-4- [methyl(7H-pyrrolo[2,3-c ]pyrimidin-4-yl)amino]cyclohexyl}methane^ maleate in dogs.
Pharmacokinetic and Pharmacodynamic Studies:
A. Pharmacokinetic Studies
/V-methyl-1-{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide has been studied as the maleate salt (Λ/- methyl-1-{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide maleate), hereinafter Compound 1 . All doses are expressed in terms of mg/kg of free base. Serial blood samples for determination of pharmacokinetics were collected following Compound 1 administration. Blood samples at 0,1 , 4, 8, and 12 hours post dose were collected via jugular venipuncture into
K2EDTA tubes and placed on ice until centrifuged. Harvested plasma was stored at approximately -20°C until analysis. .
Thirty-two beagle dogs (16 female, 16 male) were allocated to four treatment groups. Dogs in Treatment 1 received placebo capsule (0 mg/kg) orally. Dogs in Treatments 2, 3 and 4 received a combination of whole and half 3.6, 5.4, and 16 mg sized tablets orally to result in target doses of 0.6, 1.8 and 3.0 mg/kg, respectively. All dogs received a twice daily doses 0, 0.6, 1.8, and 3.0 mg/kg for weeks 1 through 6 and a single daily doses of 0, 0.6, 1.8, and 3.0 mg/kg for weeks 7 through 26. On pharmacokinetic sample collection days, dogs were fasted the previous night and fed four hours post dose administration. To calculate AUC0-24 for study days 53 and 168, steady-state was assumed and the 24 hour concentration was assumed to be equal to the 0 hour concentration, and used for the 0 hour concentration. As the actual doses of Compound 1 varied from the group target doses, the pharmacokinetic variables AUC0-T (AUC of the dosing interval), Cmax, and Cx (trough concentration) were all normalized to the group target dose.
On Day 0, AUCo-12 and Cmax increased in a dose related manner following oral administration of tablets dosed at target doses of 0.6 mg/kg, 1.8 mg/kg and 3.0 mg/kg. The increase in AUCo-12 and Cmax was dose proportional from 0.6 to 3.0 mg/kg.
Across all days and doses, there did not appear to be any systematic male/female differences in pharmacokinetic parameters. Plasma exposure increased with the number of doses following twice a day administration with a significant difference at the 0.10 level in AUCo-12 for study Day 0 in comparison to Day 21. There was a numerical decrease in the plasma exposure over the 24 hour period following the change in dosing regimen to once a day on Day 43. The Day 0 and Day 53 least- squares mean values for Cmax were not significantly different for 0.6, 1 .8, and 3.0 mg/kg. Day 53 and Day 168 the least-squares mean values for Cmax and AUC0-24 were not significantly different for 1 .8 and 3.0 mg/kg. While the Day 53 and Day 168 least- squares mean values for Cmax and AUC0-24 following 0.6 mg/kg once a day were significantly different, the CT values were not.
Other pharmacokinetic studies were completed that demonstrated following oral administration, Compound 1 was rapidly absorbed in dogs with mean maximum plasma concentrations occurring at approximately 1 hour post dose. This absorption is consistent with the observed rapid onset of pruritus reduction in both laboratory and field studies (Cosgrove, Wren et al. 2012; Fleck, Humphrey et al. 2012). Compound 1 is a low clearance compound with a moderate volume of distribution. The absolute oral bioavailability was high with a mean range of 79% to 89%. Furthermore, it can be concluded that the absorption is nearly complete based on the calculated fraction absorbed of greater than 0.9 (based on bioavailability of 85%, mean clearance of 4 mL/min/kg, and blood flow of 40 mL/min/kg). The observed increase in mean AUCo-12 from Day 0 to Day 21 of 40% was slightly greater than the expected increase of approximately 15% based on plasma elimination half-life (t½) of 4 hours. Though assuming a t½ of 4 hours, it would be predicted that steady state would be achieved by the second dose following the dose regimen change from twice daily to once daily. The similarity of the observed pharmacokinetic parameters on Day 53 (change from twice a day to once a day was on day 43) and Day 168 at 0.6 mg/kg once a day supports this conclusion.
The observed pharmacokinetic parameters of rapid oral absorption and high bioavailability are consistent with the physicochemical properties of Compound 1 . The in vitro permeability of Compound 1 was experimentally determined in a Caco-2 cell monolayer study. The permeability was high, 40.4 x 10~6 cm/sec, greater than the control for high permeability (Pfizer internal data not shown). Additionally, the solubility of Compound 1 is pH dependent with a significant drop in solubility above pH 4 down to practically insoluble by pH 5.5. The dog gastric pH has been reported to range from 1 .08 to 2.0 (Sagawa, Li et al. 2009; Mahar, Portelli et al. 2012). At this pH range and the solubility of Compound 1 (10.43 mg/mL at pH3.8) the dose for a 10 kg dog (6 mg) would fully dissolve 0.6 imL Though the estimation of the liquid volume to use for a dog is complex due to the relative small number of studies in the literature, the large size differences among individual dogs and the lack of administration of water with doses, the suggested volumes for a 10 kg dog of 9 to 20 ml. are well above what is needed for Compound 1 to be fully soluble (Martinez and Papich 2012). The solubility profile of Compound 1 is also supportive of the lack of a prandial effect. Though the pH in the fed state has been shown to spike to around pH 7, a majority of the time the pH is 2 to 4. Thus, under these conditions, Compound 1 is expected to be fully dissolved in both states, furthering supporting the observed experimental result that Compound 1 , given with or without food results in a similar oral pharmacokinetic profile. This result of similar pharmacokinetics is important for ease of administration by pet owners because dosing time does not have to be considered in respect to feed time like other AD treatments in dog.
The plasma concentration time profiles and the pharmacokinetic parameters following IV and PO administration to beagle and mongrel dogs were very similar. Although a statistical test for equivalence was not performed due to the inability to randomize beagles and mongrels between rooms, the similarity of the means and the overlap of the confidence intervals following both IV and oral administration leads to the conclusion that breed does not impact the pharmacokinetic profile. Though a formal population model was not developed all the pharmacokinetic data predicts that no clinically different pharmacokinetic profiles in client-owned dog would be different from those reported here.
The pharmacokinetic studies demonstrate that at the dose of 0.4 to 0.6 mg/kg Compound 1 exhibits rapid and nearly complete absorption, low clearance, no pharmacokinetic differences in male, female, fed, fasted, beagle and mongrel dogs, and dose proportionality. These pharmacokinetic properties are ideal for a daily or twice- daily orally administered product for the control of pruritus associated with allergic dermatitis and control of atopic dermatitis. B. Pharmacodynamic Studies:
The relationship of drug levels to pharmacodynamic effects (inhibition of cytokine function) is another factor that is important to consider when assessing potential effects of inhibiting Janus kinases. Compound 1 is a reversible inhibitor, and there is a direct relationship with drug levels and inhibition of cytokine function. Therefore, when drug levels reach IC50 levels or higher, there is a potential to significantly inhibit the function of certain cytokines, either favorably in terms of efficacy or unfavorably in terms of safety. Drug levels seen at the 0.6 mg/kg dose of compound 1 given either twice daily or once daily are shown in Figure 1.
Compound 1 inhibits cytokine receptors that share the common gamma chain
(e.g. IL-2 , IL-4R), since IC50's of representative family members range from 63-249 nM or 21-84 ng/mL, and drug levels reach those levels or higher for a significant period of time after dosing. Compound 1 also inhibits the function of a variety of cytokine receptors that share the gp130 subunit (e.g. IL-6) as well as IL-13. And finally, although not wishing to be bound by a theory, it is possible that compound 1 inhibits the function of Type I and II interferons based on the JAKs that are used by its receptor for signaling (see Figure 1 ). However, it does not appear that compound 1 inhibits cytokines from the - the IL-10 family, the IL-12 family (that share the p40 subunit), or the IL-3 family. Additionally, hormone receptors that utilize JAK2 are not substantially inhibited since drug levels do not get above IC50's for other related receptor systems utilizing JAK2 exclusively for signaling (Figure 1 ).
Collective knowledge of cytokine biology, cell types that may express JAK- dependent cytokine receptors as well as potency of compound 1 toward various classes of JAK-dependent cytokine receptors were used to help identify potential preferred dosing regimen of compound 1 (see Figure 1 ). A variety of assessments were incorporated into the dosing study to evaluate the potential for any of the identified risks as well as unanticipated risks that our Janus kinase inhibitor, compound 1 , may pose to the animal. These were contrasted with the need for adequate drug to impart efficacy.
Laboratory data supporting the identifying dosing regimen included: 1 ) inhibition (ICso's less than 249 nM or 84 ng/mL) of JAK-1 dependent cytokines (e.g. IL-2, IL-4, IL- 6, IL-13, and IL-31 ) and the ability to achieve drug levels that will inhibit these cytokines with the twice daily or once daily dosing regimen at the recommended use dose, 2) IC50's for cytokines that are exclusively dependent on JAK2 function (EPO, GM-CSF) are 4-17-fold less potent than the JAK1 -dependent cytokines evaluated and involved in allergic skin disease (IL-2, 4, 6, 13, and 31 ), 3) IC50's for other cytokines that utilize JAK2/TYK2 and not JAK1 (IL-12 and IL-23) are greater than 3000 nM (IL-12, IL-23) , 4) peak drug levels (973 nM or 328 ng/mL) observed in the margin of safety study (0.6 mg/kg dose) do not reach above the IC50's for any of the cytokines dependent on JAK2 (EPO, GM-CSF, IL-12, IL-23), and 4) significant decreases in red blood cell parameters that fall outside of normal laboratory reference ranges were not detected at the 0.6 mg/kg dose when evaluated.
The plasma concentrations that are achieved following the efficacious dose of 0.4 to 0.6 mg/kg are thought to balance safety and efficacy. The observed plasma concentrations following the twice a day regimen resulted in plasma concentrations that are greater than the JAK-1 dependent cytokine inhibitory concentrations for the entire dosing regimen. Thus, in order to fall within the preferred inhibition corridor, Compound 1 was not constantly administered twice daily for chronic use. Twice daily dosing for the first 14 days of treatment is intended to rapidly safely and effectively break the itch scratch cycle, and down-regulate the inflammatory, allergic and pruritogenic cytokine activity. Subsequent once daily dosing provides a solid margin of safety while maintaining efficacy for chronic use.

Claims

CLAIMS What is claimed is:
1 . A method for treating allergic dermatitis, atopic dermatitis, or one or more symptoms thereof in a mammal in need, which method comprises administering to the mammal a first therapeutically effective dose of a Janus Kinase (JAK) inhibitor at least twice a day for a number of days sufficient to ease or eliminate one or more clinical signs in the mammal, followed by a second therapeutically effective dose of the JAK inhibitor at a reduced frequency.
2. A method according to Claim 1 for treating pruritus associated with allergic dermatitis or pruritus associated with atopic dermatitis.
3. A method according to Claim 1 wherein the first therapeutically effective dose and the second therapeutically effective dose are administered orally.
4. A method according to Claim 1 wherein the first therapeutically effective dose is administered parenterally and the second therapeutically effective dose is administered orally.
5. A method according to any one of Claims 1 -4, wherein the JAK inhibitor is Λ/-ιη ethyl- 1 -{trans-4-[m ethyl (7/- -pyrrolo[2 , 3-d] py ri m id i n-4- yl)amino]cyclohexyl}methanesulfonamide, or a pharmaceutically acceptable salt thereof.
6. A method according to any one of claims 1 -5, wherein the first therapeutically effective dose is administered twice a day.
7. A method according to any one of claims 1 -6, wherein the first therapeutically effective dose is from about 0.4 to about 0.6 mg/kg body weight of the mammal.
8. A method according to any one of claims 1 -7, wherein the second therapeutically effective dose is administered once a day.
9. A method according to any one of claims 1 -8, wherein the second therapeutically effective dose is from about 0.4 to about 0.6 mg/kg body weight of the mammal.
10. A method according to any one of the previous claims, wherein the number of days of administration of the first therapeutically effective dose is from 1 day to 42 days, preferably 14 days.
1 1. A method for improving the therapeutic ratio of a Janus Kinase-1 (JAK-1 ) inhibitor, comprising: administering to a mammal, over a period of at least 5-days, a plurality of therapeutically effective doses of said JAK-1 inhibitor sufficient to inhibit interleukins dependent on JAK-1 ; wherein the plurality of therapeutically effective doses do not reach peak drug levels of the JAK-1 inhibitor above the IC50 for a hematopoietic cytokine.
12. A method according to claim 1 1 , wherein said interleukins are selected from the group consisting of IL-31 , IL-4, IL-2, IL-6 and IL-13.
13. A method according to claim 1 1 or 12, wherein the hematopoietic cytokines are selected from the group consisting of erythropoietin (EPO) or granulocyte colony-stimulating factor (GM-CSF).
14. A method according to any one of Claims 11 -13, wherein the JAK inhibitor is /V-methyl-1-{frans-4-[methyl(7H-pyrrolo[2,3-c ]pyrimidin-4- yl)amino]cyclohexyl}methanesulfonamide, or a pharmaceutically acceptable salt thereof.
15. A method according to any one of the previous claims, wherein the mammal is a companion animal selected from a dog and a cat.
16. A method according to Claim 15, wherein the mammal is a dog.
PCT/US2013/051015 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors WO2014015107A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
ES13742573T ES2707627T3 (en) 2012-07-20 2013-07-18 Dosage regimen for Janus kinase inhibitors (JAK)
US14/415,047 US9522151B2 (en) 2012-07-20 2013-07-18 Dosing regimen for Janus Kinase (JAK) inhibitors
IN370DEN2015 IN2015DN00370A (en) 2012-07-20 2013-07-18
NZ703152A NZ703152A (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
MX2015000871A MX360857B (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors.
AU2013292547A AU2013292547B2 (en) 2012-07-20 2013-07-18 Dosing regimen for Janus Kinase (JAK) inhibitors
KR1020157001085A KR20150028299A (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
EP13742573.2A EP2874630B1 (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
CA2878867A CA2878867C (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
SI201331328T SI2874630T1 (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
LTEP13742573.2T LT2874630T (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
RS20190051A RS58242B1 (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
BR112015000808A BR112015000808A2 (en) 2012-07-20 2013-07-18 dosage regimen for janus kinase inhibitors (jak)
CN201380038563.3A CN104470525A (en) 2012-07-20 2013-07-18 Dosage regimen for JANUS kinase (JAK)
PL13742573T PL2874630T3 (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
KR1020177007913A KR20170034949A (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors
JP2015523243A JP6022063B2 (en) 2012-07-20 2013-07-18 Administration plan of Janus kinase (JAK) inhibitor
DK13742573.2T DK2874630T3 (en) 2012-07-20 2013-07-18 Dosage regimen for Janus kinase (JAK) inhibitors
ZA2015/00134A ZA201500134B (en) 2012-07-20 2015-01-08 Dosing regimen for janus kinase (jak) inhibitors
HK15109423.1A HK1209314A1 (en) 2012-07-20 2015-09-24 Dosing regimen for janus kinase (jak) inhibitors janus (jak)
HRP20182088TT HRP20182088T1 (en) 2012-07-20 2018-12-10 Dosing regimen for janus kinase (jak) inhibitors
CY20191100203T CY1121436T1 (en) 2012-07-20 2019-02-14 DOSAGE FOR JANUS CHINESE INHIBITORS (JAK)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261674289P 2012-07-20 2012-07-20
US61/674,289 2012-07-20
US201361815803P 2013-04-25 2013-04-25
US61/815,803 2013-04-25

Publications (1)

Publication Number Publication Date
WO2014015107A1 true WO2014015107A1 (en) 2014-01-23

Family

ID=48901189

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/051015 WO2014015107A1 (en) 2012-07-20 2013-07-18 Dosing regimen for janus kinase (jak) inhibitors

Country Status (24)

Country Link
US (1) US9522151B2 (en)
EP (1) EP2874630B1 (en)
JP (2) JP6022063B2 (en)
KR (2) KR20150028299A (en)
CN (2) CN108354938A (en)
AU (1) AU2013292547B2 (en)
BR (1) BR112015000808A2 (en)
CA (1) CA2878867C (en)
CY (1) CY1121436T1 (en)
DK (1) DK2874630T3 (en)
ES (1) ES2707627T3 (en)
HK (2) HK1209314A1 (en)
HR (1) HRP20182088T1 (en)
HU (1) HUE042771T2 (en)
IN (1) IN2015DN00370A (en)
LT (1) LT2874630T (en)
MX (1) MX360857B (en)
NZ (1) NZ703152A (en)
PL (1) PL2874630T3 (en)
PT (1) PT2874630T (en)
RS (1) RS58242B1 (en)
SI (1) SI2874630T1 (en)
WO (1) WO2014015107A1 (en)
ZA (1) ZA201500134B (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9545405B2 (en) 2013-02-22 2017-01-17 Pfizer Inc. Pyrrolo[2,3-D]pyrimidine derivatives
CN107098908A (en) * 2016-02-23 2017-08-29 欣凯医药科技(上海)有限公司 A kind of preparation method and application of azolopyrimidines
US10966980B2 (en) 2014-08-12 2021-04-06 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine derivatives
EP3915989A4 (en) * 2019-01-30 2022-03-09 Gargamel (Zhuhai) Biotech Ltd. Jak inhibitor and preparation method therefor
RU2812575C2 (en) * 2019-01-30 2024-01-30 ФеликаМедБайотекнолоджи Ко., Лтд. Jak inhibitor and method of its obtaining
US11976077B2 (en) 2015-10-16 2024-05-07 Abbvie Inc. Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-α]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms therof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017143014A1 (en) * 2016-02-16 2017-08-24 Brian Kim Jak inhibitors and uses thereof
JP2023554665A (en) 2020-12-18 2023-12-28 ベーリンガー インゲルハイム アニマル ヘルス ユーエスエイ インコーポレイテッド Boron-containing pyrazole compounds, compositions containing them, methods and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020019526A1 (en) 1998-06-19 2002-02-14 Blumenkopf Todd A. Pyrrolo[2,3-d]pyrimidine compounds
WO2010020905A1 (en) * 2008-08-20 2010-02-25 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020019526A1 (en) 1998-06-19 2002-02-14 Blumenkopf Todd A. Pyrrolo[2,3-d]pyrimidine compounds
WO2010020905A1 (en) * 2008-08-20 2010-02-25 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds
US20100075996A1 (en) 2008-08-20 2010-03-25 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
"Remingtons Pharmaceutical Sciences", 1991, MACK PUB. CO.
KISSELEVA ET AL., GENE, vol. 285, 2002, pages 1
YAMAOKA ET AL., GENOME BIOLOGY, vol. 5, 2004, pages 253

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9545405B2 (en) 2013-02-22 2017-01-17 Pfizer Inc. Pyrrolo[2,3-D]pyrimidine derivatives
US9549929B2 (en) 2013-02-22 2017-01-24 Pfizer Inc. Pyrrolo[2,3-D]pyrimidine derivatives
US10966980B2 (en) 2014-08-12 2021-04-06 Pfizer Inc. Pyrrolo[2,3-d]pyrimidine derivatives
US11976077B2 (en) 2015-10-16 2024-05-07 Abbvie Inc. Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-α]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms therof
CN107098908A (en) * 2016-02-23 2017-08-29 欣凯医药科技(上海)有限公司 A kind of preparation method and application of azolopyrimidines
WO2017143990A1 (en) * 2016-02-23 2017-08-31 欣凯医药科技(上海)有限公司 Method for preparing pyrrolopyrimidine compound and application thereof
US11591333B2 (en) 2016-02-23 2023-02-28 Cinakate Pharm Tech (Shanghai) Co., Ltd. Substituted pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
EP3915989A4 (en) * 2019-01-30 2022-03-09 Gargamel (Zhuhai) Biotech Ltd. Jak inhibitor and preparation method therefor
RU2812575C2 (en) * 2019-01-30 2024-01-30 ФеликаМедБайотекнолоджи Ко., Лтд. Jak inhibitor and method of its obtaining

Also Published As

Publication number Publication date
CN108354938A (en) 2018-08-03
PT2874630T (en) 2019-01-31
MX360857B (en) 2018-11-20
JP6022063B2 (en) 2016-11-09
AU2013292547B2 (en) 2017-05-04
CA2878867A1 (en) 2014-01-23
KR20150028299A (en) 2015-03-13
EP2874630B1 (en) 2018-12-05
JP2017019816A (en) 2017-01-26
MX2015000871A (en) 2015-05-07
RS58242B1 (en) 2019-03-29
CN104470525A (en) 2015-03-25
HUE042771T2 (en) 2019-07-29
PL2874630T3 (en) 2019-04-30
KR20170034949A (en) 2017-03-29
EP2874630A1 (en) 2015-05-27
ES2707627T3 (en) 2019-04-04
US9522151B2 (en) 2016-12-20
ZA201500134B (en) 2016-08-31
US20150126535A1 (en) 2015-05-07
LT2874630T (en) 2019-02-11
NZ703152A (en) 2016-03-31
IN2015DN00370A (en) 2015-06-12
AU2013292547A1 (en) 2015-01-22
HK1209314A1 (en) 2016-04-01
JP2015522620A (en) 2015-08-06
CY1121436T1 (en) 2020-05-29
DK2874630T3 (en) 2019-01-28
SI2874630T1 (en) 2019-04-30
HRP20182088T1 (en) 2019-02-08
HK1252083A1 (en) 2019-05-17
CA2878867C (en) 2018-01-09
BR112015000808A2 (en) 2017-06-27

Similar Documents

Publication Publication Date Title
AU2013292547B2 (en) Dosing regimen for Janus Kinase (JAK) inhibitors
AU2009232276B2 (en) Antiviral drugs for treatment of arenavirus infection
EA030003B1 (en) Polycyclic carbamoylpyridone compound and pharmaceutical use thereof for treating hiv infection
JP6815728B2 (en) New use
US11318151B2 (en) Compound pharmaceutical composition for treating skin inflammatory diseases
JP2008069149A (en) Combination therapy for rheumatoid arthritis
JP2019534304A (en) Compounds for the treatment of inflammatory diseases and pharmaceutical compositions thereof
JP2024516302A (en) JAK1 pathway inhibitors for treating prurigo nodularis
US20240075038A1 (en) Application of pyrido[1,2-a]pyrimidinone analog
CA3153676A1 (en) Treatment of hidradenitis with jak inhibitors
US20230293544A1 (en) Methods, dosage regimens, and compositions for treating hidradenitis
US20230416244A1 (en) Imidazoquinoline compound having anti-inflammatory, antifugal, antiparasitic, and anticancer activity
AU2017342262A1 (en) Apilimod compositions and methods for using same in the treatment of alzheimer's disease
US20190336468A1 (en) Methods and compositions for the treatment of multiple sclerosis
CN103755562B (en) The preparation method of a kind of preparation method of lovastatin acid compound, composition, composition and application thereof

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: 13742573

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2878867

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 20157001085

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14415047

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2015523243

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/000871

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2013292547

Country of ref document: AU

Date of ref document: 20130718

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2013742573

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015000808

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112015000808

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150113