WO2022140516A1 - Certain (25)-iv-[(ls')~ i-c yan0-2-phenylethyl·]- 1,4-oxazepane-2- carboxamides for treating behcet's disease - Google Patents

Certain (25)-iv-[(ls')~ i-c yan0-2-phenylethyl·]- 1,4-oxazepane-2- carboxamides for treating behcet's disease Download PDF

Info

Publication number
WO2022140516A1
WO2022140516A1 PCT/US2021/064810 US2021064810W WO2022140516A1 WO 2022140516 A1 WO2022140516 A1 WO 2022140516A1 US 2021064810 W US2021064810 W US 2021064810W WO 2022140516 A1 WO2022140516 A1 WO 2022140516A1
Authority
WO
WIPO (PCT)
Prior art keywords
oxazepane
carboxamide
ethyl
cyano
phenyl
Prior art date
Application number
PCT/US2021/064810
Other languages
French (fr)
Inventor
Jimin Zhang
Original Assignee
Insmed Incorporated
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 Insmed Incorporated filed Critical Insmed Incorporated
Publication of WO2022140516A1 publication Critical patent/WO2022140516A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • Behpet’s Disease is as a multiorgan inflammatory disease of unknown origin, characterized by a wide clinical spectrum.
  • BD patients have been reported to present with recurrent oral ulcers, genital ulcers, uveitis and skin lesions. Baharav et al. (2006). Drug Discovery Today: Disease Models 3(1), pp. 11-14. Gastrointestinal and central nervous system involvements have also been reported. Sohn et al. (2003). International Immunopharmacology 3, pp. 713-721.
  • the death rate among BD patients is about 5%, with intestinal perforation, strokes, and a rupture of enlarged, weakened blood vessels (aneuiysms) as common causes of death.
  • Therapeutic management of BD typically depends on the clinical presentation and organ involved and consists of treating BD symptoms.
  • colchicine nonsteroidal anti-inflammatory drug (NSAID) and/or topical treatments are often prescribed.
  • NSAID nonsteroidal anti-inflammatory drug
  • topical treatments are often prescribed.
  • immunosuppressive agents such as antitumor necrosis factor alpha (TNFa) agents are used (e.g., infliximab or adalimumab).
  • BD Behçet’s Disease
  • the method comprises, in one embodiment, administering to the BD patient in need of treatment, a pharmaceutical composition comprising an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof:
  • R 2 is hydrogen, F, Cl, Br, OSO 2 C 1-3 alkyl, or C 1-3 alkyl;
  • R 3 is hydrogen, F, Cl, Br, CN, CF 3 , SO 2 C 1-3 alkyl, CONH 2 or SO 2 NR 4 R 5 , wherein R 4 and R 5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring; or
  • R 6 is C 1-3 alkyl, optionally substituted by 1, 2 or 3 F and/or optionally by OH, OC 1-3 alkyl, N(C 1-3 alkyl) 2 , cyclopropyl, or tetrahydropyran;
  • R 7 is hydrogen, F, Cl or CH 3 ;
  • X is O, S or CF 2
  • the pharmaceutical composition comprises an effective amount of (2S)-N- ⁇ (1S)-1-cyano-2-[4-(3- methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl ⁇ -1,4-oxazepane-2-carboxamide, referred to herein by its international nonproprietary name (INN), brensocatib (and formerly known as INS1007 and AZD7986), or a pharmaceutically acceptable salt thereof.
  • INN international nonproprietary name
  • brensocatib and formerly known as INS1007 and AZD7986
  • Administration routes include oral administration.
  • Administration schedules can be determined by the user of the method, e.g., a prescribing physician. In one embodiment, administration is once daily. In another embodiment, administration is twice daily. In another embodiment, administration 1 ⁇ daily, once every other day, once every third day, once every fourth day, 2 ⁇ weekly, 3 ⁇ weekly or 4 ⁇ weekly.
  • Figure 1 is a graph of surface proteinase 3 (PR3) expression (% of untreated cells, mean fluorescence intensity (MFI)) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P ⁇ 0.05 vs. the lowest brensocatib concentration.
  • PR3 surface proteinase 3
  • Figure 2 is a graph of surface proteinase 3 (PR3) expression (% positive cells) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P ⁇ 0.05 vs. untreated cells.
  • Figure 3 is a graph of neutrophil elastase (NE) activity (% of untreated control) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation.
  • PR3 surface proteinase 3
  • Figure 4 is a graph of proteinase 3 (PR3) activity (% of untreated control) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P ⁇ 0.05 vs. the lowest brensocatib concentration.
  • PR3 proteinase 3
  • Figure 5 is a graph showing the percentage of surviving BD mice during the treatment with the vehicle control (dotted line), 10 mg/kg brensocatib (thick solid line), or 30 mg/kg brensocatib (thin solid line) according to Example 2.
  • the methods provided herein employ reversible inhibitors of DPP1 in methods for treating BD.
  • Neutrophils contain four main types of granules: (i) azurophilic or primary granules, (ii) specific or secondary granules, (iii) gelatinase or tertiary granules, and (iv) secretory granules.
  • Azurophilic granules are believed to be the first to form during neutrophil maturation in the bone marrow and are characterized by the expression of related neutrophil serine proteases (NSPs): neutrophil elastase (NE), proteinase 3, and cathepsin G (CatG).
  • NSPs neutrophil serine proteases
  • NE neutrophil elastase
  • CatG cathepsin G
  • DPP1 lysosomal cysteine dipeptidyl peptidase 1
  • DPP1 is the proteinase that activates these three NSPs by removal of the N-terminal dipeptide sequences from their precursors during azurophilic granule assembly (Pham et al. (2004). J Immunol.173(12), pp.7277-7281).
  • DPP1 is broadly expressed in tissues but is highly expressed in cells of hematopoietic lineage such as neutrophils.
  • the three NSPs are abundantly secreted into the extracellular environment upon neutrophil activation at inflammatory sites, which has been reported to occur in Behçet disease pathology (see, e.g., Eksioglu-Demiralp et al. (2001). Clinical and Experimental Rheumatology 19(5), Supp.24), pp. S19-S24, incorporated by reference herein in its entirety), and are thought to act in combination with reactive oxygen species to assist in degradation of engulfed microorganisms inside phagolysosomes.
  • a fraction of the released proteases remains bound in an active form on the external surface of the plasma membrane, so that both soluble and membrane-bound NSPs can regulate the activities of a variety of biomolecules, such as chemokines, cytokines, growth factors, and cell surface receptors. Regulation is thought to occur by either converting the respective biomolecule to an active form or by degrading the biomolecule by proteolytic cleavage.
  • Secreted proteases can stimulate mucus secretion and inhibit mucociliary clearance, but also activate lymphocytes and cleave apoptotic and adhesion molecules (Bank and Ansorge (2001). J Leukoc Biol. 69, pp. 197–206; Pham (2006). Nat Rev Immunol. 6, pp.
  • the methods provided herein employ reversible inhibitors of DPP1 in methods for treating BD. Without wishing to be bound by theory, it is thought that the compounds of Formula (I), administered via the methods provided herein have beneficial effects via inhibition of activation of the three NSPs via the upstream inhibition of DPP1.
  • Inhibition of DPP1 reduces the amount of activated NSPs available for release during neutrophil degranulation.
  • inhibition of PR3 activation by the compounds of Formula (I) in turn leads to reduction of PR3 expression at the neutrophil membrane surface.
  • the decreased membrane-bound PR3 (mbPR3) in turn, might limit the targets to which PR3-specific antineutrophil cytoplasmic autoantibodies (ANCAs) can bind, and in turn, might attenuate neutrophil activation.
  • C 1-3 means a carbon group having 1, 2 or 3 carbon atoms.
  • alkyl includes both straight and branched chain alkyl groups and may be substituted or non-substituted. “Alkyl” groups include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, butyl, pentyl.
  • a pharmaceutically acceptable moiety e.g., a salt, dosage form, or excipient
  • a pharmaceutically acceptable moiety has one or more benefits that outweigh any deleterious effect that the moiety may have. Deleterious effects may include, for example, excessive toxicity, irritation, allergic response, and other problems and complications.
  • R 2 is hydrogen, F, Cl, Br, OSO 2 C 1-3 alkyl, or C 1-3 alkyl
  • R 3 is hydrogen, F, Cl, Br, CN, CF 3 , SO 2 C 1-3 alkyl, CONH 2 or SO 2 NR 4 R 5 , wherein R 4 and R 5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring
  • R 6 is C 1-3 alkyl, optionally substituted by 1, 2 or 3 F and/or optionally by OH, OC 1-3 alkyl, N(C 1- 3 alkyl) 2 , cyclopropyl, or tetrahydropyran
  • R 7 is hydrogen, F, Cl or CH 3
  • X is O, S or CF 2
  • Y is O or S
  • Q is CH
  • R 3 is hydrogen, F, Cl, Br, OSO 2 C 1-3 alkyl, or C 1-3 alkyl;
  • R 3 is hydrogen, F, Cl, Br, CN, CF 3 , SO 2 C 1-3 alkyl, CONH 2 or SO 2 NR 4 R 5 , wherein R 4 and R 5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring.
  • h 3 ydrogen, F, Cl or C 1-3 alkyl; and R is hydrogen, F, Cl, CN or SO 2 C 1-3 alkyl.
  • hydrogen, F or C 1-3 alkyl; and R 3 is hydrogen, F or CN.
  • Y is O or S;
  • Q is CH or N;
  • R 6 is C 1-3 alkyl, wherein the C 1-3 alkyl is optionally substituted by 1, 2 or 3 F and/or optionally substituted by OH, OC 1-3 alkyl, N(C 1-3 alkyl) 2 , cyclopropyl, or tetrahydropyran; and
  • R 7 is hydrogen, F, Cl or CH 3 .
  • C 1-3 alkyl wherein the C 1-3 alkyl is optionally substituted by 1, 2 or 3 F; and R 7 is hydrogen.
  • R 6 is C 1-3 alkyl, e.g., methyl, ethyl, or propyl. In still a further embodiment, R 6 is methyl.
  • R 2 is hydrogen, F, Cl, Br, OSO 2 C 1-3 alkyl or C 1-3 alkyl.
  • R 2 is hydrogen, F, Cl or C 1-3 alkyl.
  • R 2 is hydrogen, F or C 1-3 alkyl.
  • R 3 is hydrogen, F, Cl, Br, CN, CF 3 , SO 2 C 1-3 alkyl CONH 2 or SO 2 NR 4 R 5 , wherein R 4 and R 5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring.
  • R 3 is selected from hydrogen, F, Cl, CN or SO 2 C 1-3 alkyl.
  • R 3 is selected from hydrogen, F or CN.
  • R 6 is C1-3alkyl, wherein said C1-3alkyl is optionally substituted by 1, 2 or 3 F and optionally by one substituent selected from OH, OC 1-3 alkyl, N(C 1-3 alkyl) 2 , cyclopropyl, or tetrahydropyran.
  • R 6 is C 1-3 alkyl, wherein said C 1-3 alkyl is optionally substituted by 1, 2 or 3 F.
  • R 6 is methyl or ethyl.
  • R 6 is methyl.
  • R 7 is hydrogen, F, Cl or CH 3 . In a further embodiment R 7 is hydrogen.
  • the composition administered to the patient comprises an effective amount of (2S)-N- ⁇ (1S)-1-cyano-2-[4-(3-methyl-2-oxo-2,3- dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl ⁇ -1,4-oxazepane-2-carboxamide (brensocatib): or a pharmaceutically acceptable salt thereof.
  • the composition administered to the patient comprises an effective amount of brensocatib.
  • the compound of Formula (I) is: [0040] (2S)-N-[(1S)-1-Cyano-2-(4’-cyanobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide, [0041] (2S)-N- ⁇ (1S)-1-Cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl ⁇ -1,4-oxazepane-2-carboxamide, [0042] (2S)-N- ⁇ (1S)-1-Cyano-2-[4-(3,7-dimethyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl ⁇ -1,4-oxazepane-2-carboxamide, [0043] 4’-[(2S)-N-[(1
  • the BD treatment methods provided herein comprise the administration of a composition comprising an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a patient in need of BD treatment.
  • the compounds of Formula (I) and their pharmaceutically acceptable salts are inhibitors of dipeptidyl peptidase 1 (DPP1) activity.
  • the compound is brensocatib, or a pharmaceutically acceptable salt thereof.
  • Administration routes include oral administration.
  • Administration schedules can be determined by the user of the method, e.g., a prescribing physician. In one embodiment, administration is once daily. In another embodiment, administration is twice daily. In another embodiment, administration 1 ⁇ daily, once every other day, once every third day, once every fourth day, 2 ⁇ weekly, 3 ⁇ weekly or 4 ⁇ weekly. In one embodiment, the patient is administered a compound of Formula (I) 1 ⁇ daily at a dosage of 10 mg, 25 mg or 40 mg.
  • treating in one embodiment, includes one or more of the following: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in the patient that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition; (2) inhibiting the state, disorder or condition (e.g., arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); (3) relieving the condition (for example, by causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms); (4) prophylaxis against BD.
  • the BD treatable by the methods provided herein is BD that presents with one or more severe manifestations.
  • the one or more severe manifestations in one embodiment, is posterior uveitis, retinal vasculitis, vascular manifestation, neurological manifestation or a gastrointestinal (GT) manifestation.
  • GT gastrointestinal
  • Successful treatment of the BD will depend on the clinical manifestations of the disease. Typically, attenuation or elimination of a clinical or paraclinical sign of the BD will constitute effective treatment. For example, disappearance or attenuation of muco-cutaneous manifestations, arthralgia/arthritis, a neurological symptom, a GI manifestation or a vascular manifestation, in one embodiment, constitutes effective treatment. In another embodiment, prevention of death or reduction in mortality rate due to BD constitutes effective treatment.
  • a composition comprising an effective amount of a compound of Formula (I) is administered orally.
  • the compound of Formula (I) is brensocatib, or a pharmaceutically acceptable salt thereof.
  • administration is lx daily, once every other day, once every third day, once every fourth day, 2* weekly, 3* weekly or 4* weekly. In even a further embodiment, administration of the compound is once daily.
  • the dosage administered will vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated.
  • the daily dosage of the compound of the disclosure may be in the range from 0.01 micrograms per kilogram body weight (ug/kg) to 100 milligrams per kilogram body weight (mg/kg).
  • the compound of Formula (I) is administered in an oral dosage form.
  • the compound of Formula (I) is administered as a 10 mg to 50 mg dosage form, for example, a 5 mg dosage form, a 10 mg dosage form, a 15 mg dosage form, a 20 mg dosage form, a 25 mg dosage form, a 30 mg dosage form, 35 mg dosage form, a 40 mg dosage form, a 45 mg dosage form or a 50 mg dosage form.
  • the dosage form is a 25 mg or 40 mg dosage form.
  • the dosage form is administered once daily.
  • the compound is cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-l,3-benzoxazol-5-yl)phenyl]ethyl ⁇ -l,4-oxazepane- 2-carboxamide (i.e., brensocatib), or a pharmaceutically acceptable salt thereof.
  • Treating in one embodiment, is carried out over an administration period of about 3 months, about 6 months, about 9 months, about 12 months, about 15 months, about 18 months, about 21 months or about 24 months.
  • the compounds of Formula (I), or pharmaceutically acceptable salts thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the Formula (I) compound/salt (active ingredient) is in a composition comprising a pharmaceutically acceptable adjuvant(s), diluents(s) and/or carrier(s).
  • a pharmaceutically acceptable adjuvant(s), diluents(s) and/or carrier(s) Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, “Pharmaceuticals - The Science of Dosage Form Designs”, M. E. Aulton, Churchill Livingstone, 2 nd Ed. 2002, incorporated by reference herein in its entirety for all purposes.
  • the pharmaceutical composition will comprise from 0.05 to 99 %w (percent by weight), for example, from 0.05 to 80 %w, or from 0.10 to 70 %w, or from 0.10 to 50 %w, of active ingredient, all percentages by weight being based on total composition.
  • the oral dosage form is a film-coated oral tablet.
  • the dosage form is an immediate release dosage form with rapid dissolution characteristics under in vitro test conditions.
  • the oral dosage form is administered once daily.
  • the oral dosage form is administered at approximately the same time every day, e.g., prior to breakfast.
  • composition comprising an effective amount of Formula (I) is administered 2 ⁇ day.
  • composition comprising an effective amount of Formula (I) is administered once-a-week, every other day, every third day, 2 ⁇ per week, 3 ⁇ per week, 4 ⁇ per week, or 5 ⁇ per week.
  • the compound of the disclosure may be admixed with adjuvant(s), diluent(s) or carrier(s), for example, lactose, saccharose, sorbitol, mannitol; starch, for example, potato starch, corn starch or amylopectin; cellulose derivative; binder, for example, gelatine or polyvinylpyrrolidone; disintegrant, for example cellulose derivative, and/or lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, wax, paraffin, and the like, and then compressed into tablets.
  • adjuvant(s) for example, lactose, saccharose, sorbitol, mannitol
  • starch for example, potato starch, corn starch or amylopectin
  • cellulose derivative for example, gelatine or polyvinylpyrrolidone
  • disintegrant for example cellulose derivative
  • lubricant for example, magnesium stearate, calcium
  • the cores may be coated with a suitable polymer dissolved or dispersed in water or readily volatile organic solvent(s).
  • the tablet may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • the compound of the disclosure may be admixed with, for example, a vegetable oil or polyethylene glycol.
  • Hard gelatine capsules may contain granules of the compound using pharmaceutical excipients like the above-mentioned excipients for tablets.
  • liquid or semisolid formulations of the compound of the disclosure may be filled into hard gelatine capsules.
  • the composition is an oral disintegrating tablet (ODT).
  • ODTs differ from traditional tablets in that they are designed to be dissolved on the tongue rather than swallowed whole
  • the composition is an oral thin film or an oral disintegrating film (ODF).
  • ODF oral disintegrating film
  • Such formulations when placed on the tongue, hydrate via interaction with saliva, and release the active compound from the dosage form.
  • the ODF in one embodiment, contains a film-forming polymer such as hydroxypropylmethylcellulose (HPMC), hydroxypropyl cellulose (HPC), pullulan, carboxymethyl cellulose (CMC), pectin, starch, polyvinyl acetate (PVA) or sodium alginate.
  • Liquid preparations for oral application may be in the form of syrups, solutions or suspensions. Solutions, for example, may contain the compound of the disclosure, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain coloring agents, flavoring agents, saccharine and/or carboxymethylcellulose as a thickening agent. Furthermore, other excipients known to those skilled in art may be used when making formulations for oral use. [0094] A compound of Formula (I), or a pharmaceutically acceptable salt thereof, may also be administered in conjunction with a further compound used for the treatment of BD via one of the methods described herein.
  • the further compound is administered concurrently, sequentially or in admixture with a compound of Formula (I), for the treatment of BD.
  • the further compound is an NSAID.
  • the further compound is an immunosuppressive agent.
  • the immunosuppressive agent is thalidomide. In another embodiment, the immunosuppressive agent is azathioprine.
  • the further compound is colchicine. [0101] In another embodiment, the further compound is interferon alfa-2a. See e.g., Alpsoy et al. (2002). Archives of Dermatology 138, pp. 467-471. [0102] In yet another embodiment, the further compound is colchicine, levamisole hydrochloride, a corticosteroid, chlorambucil, cyclophosphamide, cyclosporine, azathioprine or thalidomide.
  • the compound of the disclosure, or a pharmaceutically acceptable salt thereof is administered concurrently or sequentially with one or more further active ingredients selected from one or more of those provided above.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof may be administered concurrently or sequentially with a further pharmaceutical composition for use as a medicament for the treatment of BD.
  • the further pharmaceutical composition may be a medicament which the patient may already be prescribed (e.g., an existing standard of care medication), and may itself be a composition comprising one or more active ingredients selected from those defined above.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof is synthesized according to the methods set forth in U.S. Patent No.
  • a compound of Formula (I) can be administered as a pharmaceutically acceptable salt.
  • a pharmaceutically acceptable salt of a compound of Formula (I) may be advantageous due to one or more of its chemical or physical properties, such as stability in differing temperatures and humidities, or a desirable solubility in H 2 O, oil, or other solvent. In some instances, a salt may be used to aid in the isolation or purification of the compound of Formula (I).
  • pharmaceutically acceptable salts include, but are not limited to, an alkali metal salt, e.g., Na or K, an alkali earth metal salt, e.g., Ca or Mg, or an organic amine salt.
  • pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid addition salts.
  • Salts and co-crystals may be characterized using well known techniques, for example X-ray powder diffraction, single crystal X-ray diffraction (for example to evaluate proton position, bond lengths or bond angles), solid state NMR, (to evaluate for example, C, N or P chemical shifts) or spectroscopic techniques (to measure for example, O-H, N-H or COOH signals and IR peak shifts resulting from hydrogen bonding).
  • X-ray powder diffraction for example to evaluate proton position, bond lengths or bond angles
  • solid state NMR to evaluate for example, C, N or P chemical shifts
  • spectroscopic techniques to measure for example, O-H, N-H or COOH signals and IR peak shifts resulting from hydrogen bonding.
  • certain compounds of Formula (I) may exist in solvated form, e.g., hydrates, including solvates of a pharmaceutically acceptable salt of a compound of Formula (I).
  • certain compounds of Formula (I) may exist as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. It is to be understood that the present disclosure encompasses all such isomeric forms.
  • Certain compounds of Formula (I) may also contain linkages (e.g., carbon-carbon bonds, carbon- nitrogen bonds such as amide bonds) wherein bond rotation is restricted about that particular linkage, e.g., restriction resulting from the presence of a ring bond or double bond.
  • Certain compound of Formula (I) may also contain multiple tautomeric forms. It is to be understood that the present disclosure encompasses all such tautomeric forms. Stereoisomers may be separated using conventional techniques, e.g., chromatography or fractional crystallization, or the stereoisomers may be made by stereoselective synthesis. [0114] In a further embodiment, the compounds of Formula (I) encompass any isotopically-labeled (or “radio-labelled”) derivatives of a compound of Formula (I).
  • Such a derivative is a derivative of a compound of Formula (I) wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature.
  • radionuclides that may be incorporated include 2 H (also written as “D” for deuterium).
  • a compound of Formula (I) is provided where one or more hydrogen atoms are replaced by one or more deuterium atoms; and the deuterated compound is used in one of the methods provided herein for treating BD.
  • the compounds of Formula (I) may be administered in the form of a prodrug which is broken down in the human or animal body to give rise to a compound of the Formula (I).
  • prodrugs include in vivo hydrolysable esters of a compound of the Formula (I).
  • An in vivo hydrolysable (or cleavable) ester of a compound of Formula (I) that contains a carboxy or a hydroxy group is, for example, a pharmaceutically acceptable ester which is hydrolyzed in the human or animal body to produce the parent acid or alcohol.
  • ester prodrugs derivatives see: Curr. Drug. Metab. 2003, 4, 461, incorporated by reference herein in its entirety for all purposes.
  • Various other forms of prodrugs are known in the art, and can be used in the methods provided herein.
  • AllCells Primary bone marrow- or umbilical cord blood-derived CD34 + neutrophil progenitor cells (AllCells; Alameda, CA) were cultured for 7 days in STEMSPAN SFEM media (Stemcell Technologies; Tukwila, WA) supplemented with recombinant human Stem Cell Factor (Peprotech; Rocky Hill, NJ) and recombinant human IL-3 (Peprotech).
  • the cells were differentiated in culture for another 7 days in STEMSPAN SFEM media (Stemcell Technologies) with recombinant human Granulocyte Colony Stimulating Factor (Peprotech), plus increasing concentrations of brensocatib (0 – ⁇ ⁇ 0 ⁇ ⁇ $W ⁇ WKH ⁇ HQG ⁇ RI ⁇ WKH ⁇ differentiation/treatment period, cells were split into two aliquots for analysis. [0120] The first aliquot of differentiated neutrophils was harvested for flow cytometry to quantify PR3 expression on the surface of neutrophils. After harvest, the cells were resuspended in FACS staining buffer and Fc receptors were blocked for 30 min at 4 °C. Cells were stained for 30 min.
  • Peptide substrates for NE N-Methoxysuccinyl-Ala- Ala-Pro-Val-7-amido-4-methylcoumarin; Sigma-Aldrich; St. Louis, MO
  • PR3 aminobenzoyl-Val-Ala-Asp-Cys-Ala-Asp-Gln-Lys(2,4-dinitrophenyl); GenScript; Piscataway, NJ
  • CatG N-Succinyl-Ala-Ala-Pro-Phe p-nitroanilide; Sigma
  • fluorescence or absorbance was quantified using a Synergy microplate reader (BioTek; Winooski, VT).
  • NSP inhibitors were used to test the specificity of enzymatic assays, including elastase inhibitor (Abcam) for NE, sivelestat (Abcam) for PR3, and cathepsin G inhibitor I (Cayman Chemical; Ann Arbor, MI) for CatG.
  • Abcam elastase inhibitor
  • sivelestat Abcam
  • cathepsin G inhibitor I Cayman Chemical; Ann Arbor, MI
  • a portion of cell lysate samples were retained for protein quantification using a PierceTM BCA Protein Assay Kit (Thermo Fisher) according to the manufacturer’s instructions.
  • the NSP activities were normalized to cell lysate protein concentrations and the final data were fit with nonlinear, four-parameter, logistic regression analysis by using GraphPad Prism software (GraphPad Software Inc.; La Jolla, CA).
  • neutrophils were harvested, stained for 30 min with either a mouse anti-PR3 monoclonal antibody (Abcam; Cambridge, MA) or a mouse monoclonal IgG isotype control (Abcam), followed by a Cy5-conjugated goat anti-mouse IgG secondary antibody (Abcam), fixed in 4% formaldehyde, and mounted to glass slides with ProLongTM Diamond Antifade Mountant with DAPI (Thermo Fisher; Waltham, MA). Samples were imaged with an Axio fluorescence microscope (Zeiss; Oberkochen, Germany) using constant settings while imaging samples from different treatment conditions.
  • Example 2 Therapeutic effects of brensocatib in a HSV model of BD
  • a herpes simplex virus (HSV)-induced BD ICR mouse model was used to investigate the therapeutic effects of brensocatib.
  • ICR mice were infected with HSV to induce ulceration of mouth, genital, and skin, eye involvement, and other disease symptoms of BD in the mice, followed by treatment with brensocatib at two different doses (i.e., 10 mg brensocatib/kg body weight per mouse or 30 mg brensocatib/kg body weight per mouse) or a vehicle control.
  • BD symptom severity and mortality were compared between the brensocatib-treated groups and the vehicle-treated control group.
  • mice were infected with HSV as described previously (Shim et al., Cytotherapy 13:835-847 (2011); Sohn et al., Eur J Dermatol.8(1):21-3 (1998); each of which is incorporated by reference herein in its entirety for all purposes). Briefly, earlobes of the mice were scratched with a needle, then inoculated with ⁇ / ⁇ of 1.0 x 10 6 plaque-forming units (pfu)/mL of HSV type l (F strain) solution. Ten days later, a second inoculation was performed using the same method.
  • mice were anesthetized by intraperitoneal injection with 2,2,2-tribromo ethanol (240 mg/kg). After viral inoculation, hydrocortisone (150 ⁇ g/mouse) was intraperitoneally injected once a day for 5 days. The mice were kept in a conventional temperature- and light-controlled room (20-22 °C, 12 hr. light cycle starting at 8:00 a.m.). The mice had free access to food and water, and were handled in accordance with a protocol approved by an institutional animal care committee. [0128] Symptoms of BD were confirmed by follow-up for 5 to 16 weeks after the second virus inoculation.
  • mice Several symptoms, including oral ulcers, genital ulcers, erythema, skin pustules, skin ulcers, arthritis, diarrhea, red-eye, loss of balance, and facial edema, were observed in the mice after HSV inoculation. Oral, genital, skin ulcers, and eye symptoms were classified as major symptoms. Arthritis, intestinal ulceration, and neurological involvement were considered as minor symptoms. Mice with at least one major symptom and at least one minor symptom were classified as having BD. Each symptom was scored as one point. The sum of points was used to determine the severity of BD using 2006 BD Current Activity Form prepared by the International Society for Behçet’s Disease The disappearance of symptoms or a reduction in skin lesion size of 20% or more indicated improvement of BD.
  • Table 1 shows that, as compared to the vehicle control, brensocatib treatment at both doses resulted in higher percentages of HSV-induced BD mice with improvement in BD symptoms, no change in BD symptoms, and/or a combination of the two, at 2 weeks and 4 weeks of the administration period, based on the disease severity scores. Moreover, brensocatib treatment at the 10 mg/kg dose was more effective than at the 30 mg/kg dose in improving the BD symptoms, consistent with their effects on the reduction in mortality. Table 1.
  • Example 3 Prophylactic effects of brensocatib in a HSV model of BD
  • HSV-induced BD occurrence rate in C57BL/6 mice is estimated to be about 40%, as compared to a 10-15% occurrence rate in ICR mice used in Example 2.
  • C57BL/6 mice in groups of about 30 each are treated with brensocatib at three different doses, i.e., 3 mg/kg, 10 mg/kg, and 30 mg/kg per day, or a vehicle control solution containing tartaric acid (as described in Example 2), via oral administration twice per day, seven days prior to a first inoculation of HSV performed as described in Example 2. Ten days later a second HSV inoculation is performed also as described in Example 2. After the first inoculation of HSV, brensocatib or vehicle treatment is performed 2 times per day for 7 days, and thereafter once a day for the next 16 weeks.

Abstract

The present disclosure relates to methods for treating Behçet's Disease with compositions comprising an effective amount of certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamide compounds of Formula (I), including pharmaceutically acceptable salts thereof that inhibit dipeptidyl peptidase 1 (DPP1) activity. In one embodiment, the compound of Formula (I) is (2S)-N-{(1S)-1-cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide (brensocatib).

Description

CERTAIN (2S)-A-[(15)-l-CYANO-2-PHENYLETHYL]-l,4-OXAZEPANE-2- CARBOXAMIDES FOR TREATING BEHQET’S DISEASE
CROSS REFERENCE TO RELATED APPLICATION
[0001] This application claims priority from U.S. Provisional Application Serial No. 63/129, 133, filed December 22, 2020, the disclosure of which is incorporated by reference herein in its entirety.
BACKGROUND OF THE INVENTION
[0002] Behpet’s Disease (BD) is as a multiorgan inflammatory disease of unknown origin, characterized by a wide clinical spectrum. For example, BD patients have been reported to present with recurrent oral ulcers, genital ulcers, uveitis and skin lesions. Baharav et al. (2006). Drug Discovery Today: Disease Models 3(1), pp. 11-14. Gastrointestinal and central nervous system involvements have also been reported. Sohn et al. (2003). International Immunopharmacology 3, pp. 713-721. Pathologically, patients with BD present with systemic necrotizing vasculitis of small and large vessels, arthritis and a positive pathergy test.. The death rate among BD patients is about 5%, with intestinal perforation, strokes, and a rupture of enlarged, weakened blood vessels (aneuiysms) as common causes of death.
[0003] Therapeutic management of BD typically depends on the clinical presentation and organ involved and consists of treating BD symptoms. For example, for patients presenting mucocutaneous and joint involvement, colchicine, nonsteroidal anti-inflammatory drug (NSAID) and/or topical treatments are often prescribed. Vallet et al. (2015). Journal of Autoimmunity 62, pp. 67-74. For severe symptoms, immunosuppressive agents such as antitumor necrosis factor alpha (TNFa) agents are used (e.g., infliximab or adalimumab).
[0004] Although studies have suggested viral involvement in BD, including herpes simplex vims (HSV) involvement, treatment with the anti-viral acyclovir has not been shown to significantly alleviate the frequency of clinical features of BD. Sohn et al. (2003). International Immunopharmacology 3, pp. 713-721.
[0005] Because current drugs are not totally effective in controlling BD symptoms, let alone effective in treating the cause of BD, and have been associated with significant adverse events, a need exists for new and improved BD treatments. The present invention addresses this and other needs. SUMMARY OF THE INVENTION [0006] In one aspect, a method is provided for treating a Behçet’s Disease (BD) patient in need thereof. The method comprises, in one embodiment, administering to the BD patient in need of treatment, a pharmaceutical composition comprising an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000004_0001
R2 is hydrogen, F, Cl, Br, OSO2C1-3alkyl, or C1-3alkyl; R3 is hydrogen, F, Cl, Br, CN, CF3, SO2C1-3alkyl, CONH2 or SO2NR4R5, wherein R4 and R5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring; or R6 is C1-3alkyl, optionally substituted by 1, 2 or 3 F and/or optionally by OH, OC1-3alkyl, N(C1-3alkyl)2, cyclopropyl, or tetrahydropyran; R7 is hydrogen, F, Cl or CH3; X is O, S or CF2; Y is O or S; and Q is CH or N. [0007] In one embodiment of the method for treating BD in a patient in need thereof, the pharmaceutical composition comprises an effective amount of (2S)-N-{(1S)-1-cyano-2-[4-(3- methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, referred to herein by its international nonproprietary name (INN), brensocatib (and formerly known as INS1007 and AZD7986),
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof. In the present application, brensocatib and INS1007 are used interchangeably and refer to (2S)-N-{(1S)-1-cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide. [0008] Administration routes include oral administration. Administration schedules can be determined by the user of the method, e.g., a prescribing physician. In one embodiment, administration is once daily. In another embodiment, administration is twice daily. In another embodiment, administration 1× daily, once every other day, once every third day, once every fourth day, 2× weekly, 3× weekly or 4× weekly. BRIEF DESCRIPTION OF THE FIGURES [0009] Figure 1 is a graph of surface proteinase 3 (PR3) expression (% of untreated cells, mean fluorescence intensity (MFI)) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P < 0.05 vs. the lowest brensocatib concentration. [0010] Figure 2 is a graph of surface proteinase 3 (PR3) expression (% positive cells) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P < 0.05 vs. untreated cells. [0011] Figure 3 is a graph of neutrophil elastase (NE) activity (% of untreated control) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P < 0.05 vs. the lowest brensocatib concentration. [0012] Figure 4 is a graph of proteinase 3 (PR3) activity (% of untreated control) in neutrophils derived from either bone marrow (BM) or umbilical cord (UC) blood stem cells as a function of brensocatib concentration according to Example 1. Brensocatib was added in the cell culture medium during neutrophil differentiation. *P < 0.05 vs. the lowest brensocatib concentration. [0013] Figure 5 is a graph showing the percentage of surviving BD mice during the treatment with the vehicle control (dotted line), 10 mg/kg brensocatib (thick solid line), or 30 mg/kg brensocatib (thin solid line) according to Example 2. DETAILED DESCRIPTION OF THE INVENTION [0014] The methods provided herein employ reversible inhibitors of DPP1 in methods for treating BD. [0015] Neutrophils contain four main types of granules: (i) azurophilic or primary granules, (ii) specific or secondary granules, (iii) gelatinase or tertiary granules, and (iv) secretory granules. Azurophilic granules are believed to be the first to form during neutrophil maturation in the bone marrow and are characterized by the expression of related neutrophil serine proteases (NSPs): neutrophil elastase (NE), proteinase 3, and cathepsin G (CatG). The lysosomal cysteine dipeptidyl peptidase 1 (DPP1) is the proteinase that activates these three NSPs by removal of the N-terminal dipeptide sequences from their precursors during azurophilic granule assembly (Pham et al. (2004). J Immunol.173(12), pp.7277-7281). DPP1 is broadly expressed in tissues but is highly expressed in cells of hematopoietic lineage such as neutrophils. [0016] The three NSPs are abundantly secreted into the extracellular environment upon neutrophil activation at inflammatory sites, which has been reported to occur in Behçet disease pathology (see, e.g., Eksioglu-Demiralp et al. (2001). Clinical and Experimental Rheumatology 19(5), Supp.24), pp. S19-S24, incorporated by reference herein in its entirety), and are thought to act in combination with reactive oxygen species to assist in degradation of engulfed microorganisms inside phagolysosomes. A fraction of the released proteases remains bound in an active form on the external surface of the plasma membrane, so that both soluble and membrane-bound NSPs can regulate the activities of a variety of biomolecules, such as chemokines, cytokines, growth factors, and cell surface receptors. Regulation is thought to occur by either converting the respective biomolecule to an active form or by degrading the biomolecule by proteolytic cleavage. Secreted proteases can stimulate mucus secretion and inhibit mucociliary clearance, but also activate lymphocytes and cleave apoptotic and adhesion molecules (Bank and Ansorge (2001). J Leukoc Biol. 69, pp. 197–206; Pham (2006). Nat Rev Immunol. 6, pp. 541–550; Meyer-Hoffert (2009). Front Biosci. 14, pp. 3409–3418; Voynow et al. (2004). Am J Physiol Lung Cell Mol Physiol.287, pp. L1293-302; the disclosure of each of which is incorporated by reference in its entirety for all purposes). [0017] The methods provided herein employ reversible inhibitors of DPP1 in methods for treating BD. Without wishing to be bound by theory, it is thought that the compounds of Formula (I), administered via the methods provided herein have beneficial effects via inhibition of activation of the three NSPs via the upstream inhibition of DPP1. Inhibition of DPP1, without wishing to be bound by theory, reduces the amount of activated NSPs available for release during neutrophil degranulation. Moreover, in a small population of Behcet’s Disease patients whose pathogenesis is thought to be driven by PR3-reactive ANCAs, inhibition of PR3 activation by the compounds of Formula (I) in turn leads to reduction of PR3 expression at the neutrophil membrane surface. The decreased membrane-bound PR3 (mbPR3) in turn, might limit the targets to which PR3-specific antineutrophil cytoplasmic autoantibodies (ANCAs) can bind, and in turn, might attenuate neutrophil activation. In addition, in a small population of Behcet’s Disease patients whose pathogenesis is thought to be driven by CatG-reactive ANCAs, similar mechanism of action to the PR3 autoantigen by the compounds of Formula (I) may happen and thus the compounds have potential therapeutic effect for those patients. [0018] As used herein, “C1-3” means a carbon group having 1, 2 or 3 carbon atoms. [0019] The term “alkyl”, unless otherwise noted, includes both straight and branched chain alkyl groups and may be substituted or non-substituted. “Alkyl” groups include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, butyl, pentyl. [0020] The term “pharmaceutically acceptable”, unless otherwise noted, is used to characterize a moiety (e.g., a salt, dosage form, or excipient) as being appropriate for use in accordance with sound medical judgment. In general, a pharmaceutically acceptable moiety has one or more benefits that outweigh any deleterious effect that the moiety may have. Deleterious effects may include, for example, excessive toxicity, irritation, allergic response, and other problems and complications. [0021] Provided herein are methods for treating BD patients via administration of a pharmaceutical composition comprising an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000008_0001
R2 is hydrogen, F, Cl, Br, OSO2C1-3alkyl, or C1-3alkyl; R3 is hydrogen, F, Cl, Br, CN, CF3, SO2C1-3alkyl, CONH2 or SO2NR4R5, wherein R4 and R5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring; or R6 is C1-3alkyl, optionally substituted by 1, 2 or 3 F and/or optionally by OH, OC1-3alkyl, N(C1- 3alkyl)2, cyclopropyl, or tetrahydropyran; R7 is hydrogen, F, Cl or CH3; X is O, S or CF2; Y is O or S; and Q is CH or N. [0022] In one embodiment
Figure imgf000008_0002
hydrogen, F, Cl, Br, OSO2C1-3alkyl, or C1-3alkyl; R3 is hydrogen, F, Cl, Br, CN, CF3, SO2C1-3alkyl, CONH2 or SO2NR4R5, wherein R4 and R5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring. [0023] In a further embodiment, h 3
Figure imgf000009_0001
ydrogen, F, Cl or C1-3alkyl; and R is hydrogen, F, Cl, CN or SO2C1-3alkyl. [0024] In still a further embodiment,
Figure imgf000009_0002
hydrogen, F or C1-3alkyl; and R3 is hydrogen, F or CN.
Figure imgf000009_0003
is O, S or CF2; Y is O or S; Q is CH or N; R6 is C1-3alkyl, wherein the C1-3alkyl is optionally substituted by 1, 2 or 3 F and/or optionally substituted by OH, OC1-3alkyl, N(C1-3alkyl)2, cyclopropyl, or tetrahydropyran; and R7 is hydrogen, F, Cl or CH3. In a further embodiment,
Figure imgf000009_0004
CF2; Y is O or S; R6 is C1-3alkyl, optionally substituted by 1, 2 or 3 F and optionally substituted by OH, OC1-3alkyl, N(C1-3alkyl)2, cyclopropyl, or tetrahydropyran; and R7 is hydrogen, F, Cl or CH3. In a further embodiment,
Figure imgf000010_0001
[0027] In another embodiment,
Figure imgf000010_0002
C1-3alkyl, wherein the C 1-3alkyl is optionally substituted by 1, 2 or 3 F; and R 7 is hydrogen, F, Cl or CH3. [0028] In another embodiment,
Figure imgf000010_0003
C1-3alkyl, wherein the C1-3alkyl is optionally substituted by 1, 2 or 3 F; and R7 is hydrogen. In a further embodiment, R6 is C1-3alkyl, e.g., methyl, ethyl, or propyl. In still a further embodiment, R6 is methyl. [0029] In one embodiment, R2 is hydrogen, F, Cl, Br, OSO2C1-3alkyl or C1-3alkyl. [0030] In a further embodiment, R2 is hydrogen, F, Cl or C1-3alkyl. [0031] In still a further embodiment, R2 is hydrogen, F or C1-3alkyl. [0032] In one embodiment, R3 is hydrogen, F, Cl, Br, CN, CF3, SO2C1-3alkyl CONH2 or SO2NR4R5, wherein R4 and R5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring. [0033] In a further embodiment, R3 is selected from hydrogen, F, Cl, CN or SO2C1-3alkyl. [0034] In still a further embodiment, R3 is selected from hydrogen, F or CN. [0035] In one embodiment, R6 is C1-3alkyl, wherein said C1-3alkyl is optionally substituted by 1, 2 or 3 F and optionally by one substituent selected from OH, OC1-3alkyl, N(C1-3alkyl)2, cyclopropyl, or tetrahydropyran. [0036] In a further embodiment, R6 is C1-3alkyl, wherein said C1-3alkyl is optionally substituted by 1, 2 or 3 F. In still a further embodiment, R6 is methyl or ethyl. In still a further embodiment, R6 is methyl. [0037] In one embodiment, R7 is hydrogen, F, Cl or CH3. In a further embodiment R7 is hydrogen. [0038] In one embodiment of the methods provided herein, the composition administered to the patient comprises an effective amount of (2S)-N-{(1S)-1-cyano-2-[4-(3-methyl-2-oxo-2,3- dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide (brensocatib): or a pharmaceutically acceptable salt thereof. In a further
Figure imgf000011_0001
embodiment, the composition administered to the patient comprises an effective amount of brensocatib. [0039] In one embodiment, the compound of Formula (I) is: [0040] (2S)-N-[(1S)-1-Cyano-2-(4’-cyanobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide, [0041] (2S)-N-{(1S)-1-Cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0042] (2S)-N-{(1S)-1-Cyano-2-[4-(3,7-dimethyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0043] 4’-[(2S)-2-Cyano-2-{[(2S)-1,4-oxazepan-2-ylcarbonyl]amino}ethyl]biphenyl-3-yl methanesulfonate, [0044] (2S)-N-{(1S)-1-Cyano-2-[4-(3-methyl-1,2-benzoxazol-5-yl)phenyl]ethyl}-1,4- oxazepane-2-carboxamide, [0045] (2S)-N-{(1S)-1-Cyano-2-[4’-(trifluoromethyl)biphenyl-4-yl]ethyl}-1,4-oxazepane-2- carboxamide, [0046] (2S)-N-[(1S)-1-Cyano-2-(3’,4’-difluorobiphenyl-4-yl)ethyl]-1,4-oxazepane-2- carboxamide, [0047] (2S)-N-{(1S)-1-Cyano-2-[4-(6-cyanopyridin-3-yl)phenyl]ethyl}-1,4-oxazepane-2- carboxamide, [0048] (2S)-N-{(1S)-1-Cyano-2-[4-(4-methyl-3-oxo-3,4-dihydro-2H-1,4-benzothiazin-6- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0049] (2S)-N-{(1S)-1-Cyano-2-[4-(3-ethyl-7-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0050] (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2-hydroxy-2-methylpropyl)-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0051] (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2,2-difluoroethyl)-7-fluoro-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0052] (2S)-N-[(1S)-1-Cyano-2-(4-{3-[2-(dimethylamino)ethyl]-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl}phenyl)ethyl]-1,4-oxazepane-2-carboxamide, [0053] (2S)-N-{(1S)-1-Cyano-2-[4-(3,3-difluoro-1-methyl-2-oxo-2,3-dihydro-1H-indol-6- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0054] (2S)-N-{(1S)-1-Cyano-2-[4-(7-fluoro-3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0055] (2S)-N-{(1S)-1-Cyano-2-[4-(3-ethyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0056] (2S)-N-[(1S)-1-Cyano-2-{4-[3-(cyclopropylmethyl)-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0057] (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2-methoxyethyl)-2-oxo-2,3-dihydro-1,3-benzothiazol- 5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0058] (2S)-N-[(1S)-1-Cyano-2-{4-[2-oxo-3-(propan-2-yl)-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0059] (2S)-N-{(1S)-1-Cyano-2-[4-(4-methyl-3-oxo-3,4-dihydro-2H-1,4-benzoxazin-6- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0060] (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2-methoxyethyl)-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0061] (2S)-N-{(1S)-1-Cyano-2-[4-(5-cyanothiophen-2-yl)phenyl]ethyl}-1,4-oxazepane-2- carboxamide, [0062] (2S)-N-[(1S)-2-(4’-Carbamoyl-3'-fluorobiphenyl-4-yl)-1-cyanoethyl]-1,4-oxazepane- 2-carboxamide, [0063] (2S)-N-{(1S)-1-Cyano-2-[4-(1-methyl-2-oxo-1,2-dihydroquinolin-7-yl)phenyl]ethyl}- 1,4-oxazepane-2-carboxamide, [0064] (2S)-N-[(1S)-1-Cyano-2-{4-[2-oxo-3-(tetrahydro-2H-pyran-4-ylmethyl)-2,3-dihydro- 1,3-benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0065] (2S)-N-{(1S)-2-[4-(7-Chloro-3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]-1-cyanoethyl}-1,4-oxazepane-2-carboxamide, [0066] (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2,2-difluoroethyl)-2-oxo-2,3-dihydro-1,3-benzoxazol- 5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0067] (2S)-N-[(1S)-1-Cyano-2-{4-[2-oxo-3-(2,2,2-trifluoroethyl)-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide, [0068] (2S)-N-{(1S)-1-Cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzothiazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide, [0069] (2S)-N-{(1S)-1-Cyano-2-[4’-(methylsulfonyl)biphenyl-4-yl]ethyl}-1,4-oxazepane-2- carboxamide, [0070] (2S)-N-{(1S)-2-[4’-(Azetidin-1-ylsulfonyl)biphenyl-4-yl]-1-cyanoethyl}-1,4- oxazepane-2-carboxamide, [0071] (2S)-N-[(1S)-1-Cyano-2-(4’-fluorobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide, [0072] (2S)-N-{(1S)-2-[4-(1,3-Benzothiazol-5-yl)phenyl]-1-cyanoethyl}-1,4-oxazepane-2- carboxamide, or [0073] (2S)-N-[(1S)-1-Cyano-2-(4’-cyanobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide, [0074] or a pharmaceutically acceptable salt of one of the foregoing compounds. [0075] Formula (I), its subgenera, and brensocatib, as well as methods of making the same, are disclosed in U.S. Patent No. 9,522,894, the disclosure of which is incorporated by reference in its entirety for all purposes. [0076] The BD treatment methods provided herein comprise the administration of a composition comprising an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a patient in need of BD treatment. The compounds of Formula (I) and their pharmaceutically acceptable salts are inhibitors of dipeptidyl peptidase 1 (DPP1) activity. In one embodiment, the compound is brensocatib, or a pharmaceutically acceptable salt thereof. [0077] Administration routes include oral administration. Administration schedules can be determined by the user of the method, e.g., a prescribing physician. In one embodiment, administration is once daily. In another embodiment, administration is twice daily. In another embodiment, administration 1× daily, once every other day, once every third day, once every fourth day, 2× weekly, 3× weekly or 4× weekly. In one embodiment, the patient is administered a compound of Formula (I) 1× daily at a dosage of 10 mg, 25 mg or 40 mg. [0078] The term “treating” in one embodiment, includes one or more of the following: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in the patient that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition; (2) inhibiting the state, disorder or condition (e.g., arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); (3) relieving the condition (for example, by causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms); (4) prophylaxis against BD. [0079] The BD treatable by the methods provided herein, in one embodiment, is BD that presents with one or more severe manifestations. The one or more severe manifestations, in one embodiment, is posterior uveitis, retinal vasculitis, vascular manifestation, neurological manifestation or a gastrointestinal (GT) manifestation.
[0080] Successful treatment of the BD will depend on the clinical manifestations of the disease. Typically, attenuation or elimination of a clinical or paraclinical sign of the BD will constitute effective treatment. For example, disappearance or attenuation of muco-cutaneous manifestations, arthralgia/arthritis, a neurological symptom, a GI manifestation or a vascular manifestation, in one embodiment, constitutes effective treatment. In another embodiment, prevention of death or reduction in mortality rate due to BD constitutes effective treatment.
[0081] In a further embodiment, a composition comprising an effective amount of a compound of Formula (I) is administered orally. In a further embodiment, the compound of Formula (I) is brensocatib, or a pharmaceutically acceptable salt thereof. In yet a further embodiment, administration is lx daily, once every other day, once every third day, once every fourth day, 2* weekly, 3* weekly or 4* weekly. In even a further embodiment, administration of the compound is once daily.
[0082] The dosage administered will vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. In one embodiment, if the compound is administered orally, then the daily dosage of the compound of the disclosure may be in the range from 0.01 micrograms per kilogram body weight (ug/kg) to 100 milligrams per kilogram body weight (mg/kg).
[0083] In one embodiment, the compound of Formula (I) is administered in an oral dosage form. In a further embodiment, the compound of Formula (I) is administered as a 10 mg to 50 mg dosage form, for example, a 5 mg dosage form, a 10 mg dosage form, a 15 mg dosage form, a 20 mg dosage form, a 25 mg dosage form, a 30 mg dosage form, 35 mg dosage form, a 40 mg dosage form, a 45 mg dosage form or a 50 mg dosage form. In a further embodiment, the dosage form is a 25 mg or 40 mg dosage form. In a further embodiment, the dosage form is administered once daily. In even a further embodiment, the compound is
Figure imgf000015_0001
cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-l,3-benzoxazol-5-yl)phenyl]ethyl}-l,4-oxazepane- 2-carboxamide (i.e., brensocatib), or a pharmaceutically acceptable salt thereof. [0084] Treating, in one embodiment, is carried out over an administration period of about 3 months, about 6 months, about 9 months, about 12 months, about 15 months, about 18 months, about 21 months or about 24 months. [0085] The compounds of Formula (I), or pharmaceutically acceptable salts thereof, may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the Formula (I) compound/salt (active ingredient) is in a composition comprising a pharmaceutically acceptable adjuvant(s), diluents(s) and/or carrier(s). Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, “Pharmaceuticals - The Science of Dosage Form Designs”, M. E. Aulton, Churchill Livingstone, 2 nd Ed. 2002, incorporated by reference herein in its entirety for all purposes. [0086] Depending on the mode of administration, the pharmaceutical composition will comprise from 0.05 to 99 %w (percent by weight), for example, from 0.05 to 80 %w, or from 0.10 to 70 %w, or from 0.10 to 50 %w, of active ingredient, all percentages by weight being based on total composition. [0087] In one oral administration embodiment, the oral dosage form is a film-coated oral tablet. In a further embodiment, the dosage form is an immediate release dosage form with rapid dissolution characteristics under in vitro test conditions. [0088] In one embodiment, the oral dosage form is administered once daily. In a further embodiment, the oral dosage form is administered at approximately the same time every day, e.g., prior to breakfast. In another embodiment, the composition comprising an effective amount of Formula (I) is administered 2× day. In yet another embodiment, the composition comprising an effective amount of Formula (I) is administered once-a-week, every other day, every third day, 2× per week, 3× per week, 4× per week, or 5× per week. [0089] For oral administration the compound of the disclosure may be admixed with adjuvant(s), diluent(s) or carrier(s), for example, lactose, saccharose, sorbitol, mannitol; starch, for example, potato starch, corn starch or amylopectin; cellulose derivative; binder, for example, gelatine or polyvinylpyrrolidone; disintegrant, for example cellulose derivative, and/or lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a suitable polymer dissolved or dispersed in water or readily volatile organic solvent(s). Alternatively, the tablet may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide. [0090] For the preparation of soft gelatine capsules, the compound of the disclosure may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatine capsules may contain granules of the compound using pharmaceutical excipients like the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the disclosure may be filled into hard gelatine capsules. [0091] In one embodiment, the composition is an oral disintegrating tablet (ODT). ODTs differ from traditional tablets in that they are designed to be dissolved on the tongue rather than swallowed whole [0092] In one embodiment, the composition is an oral thin film or an oral disintegrating film (ODF). Such formulations, when placed on the tongue, hydrate via interaction with saliva, and release the active compound from the dosage form. The ODF, in one embodiment, contains a film-forming polymer such as hydroxypropylmethylcellulose (HPMC), hydroxypropyl cellulose (HPC), pullulan, carboxymethyl cellulose (CMC), pectin, starch, polyvinyl acetate (PVA) or sodium alginate. [0093] Liquid preparations for oral application may be in the form of syrups, solutions or suspensions. Solutions, for example, may contain the compound of the disclosure, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain coloring agents, flavoring agents, saccharine and/or carboxymethylcellulose as a thickening agent. Furthermore, other excipients known to those skilled in art may be used when making formulations for oral use. [0094] A compound of Formula (I), or a pharmaceutically acceptable salt thereof, may also be administered in conjunction with a further compound used for the treatment of BD via one of the methods described herein. [0095] The further compound is administered concurrently, sequentially or in admixture with a compound of Formula (I), for the treatment of BD. [0096] In one embodiment, the further compound is an NSAID. [0097] In one embodiment, the further compound is an immunosuppressive agent. [0098] In a further embodiment, the immunosuppresVLYHαDJHQWαLVα71)ĮαLQKLELWRUαα α,QαHYHQαDα IXUWKHUαHPERGLPHQWααWKHα71)ĮαLQKLELWRUαLVαLQIliximab, adalimumab or etanercept. [0099] In one embodiment, the immunosuppressive agent is thalidomide. In another embodiment, the immunosuppressive agent is azathioprine. [0100] In another embodiment, the further compound is colchicine. [0101] In another embodiment, the further compound is interferon alfa-2a. See e.g., Alpsoy et al. (2002). Archives of Dermatology 138, pp. 467-471. [0102] In yet another embodiment, the further compound is colchicine, levamisole hydrochloride, a corticosteroid, chlorambucil, cyclophosphamide, cyclosporine, azathioprine or thalidomide. [0103] In one combination therapy embodiment, the compound of the disclosure, or a pharmaceutically acceptable salt thereof, is administered concurrently or sequentially with one or more further active ingredients selected from one or more of those provided above. For example, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, may be administered concurrently or sequentially with a further pharmaceutical composition for use as a medicament for the treatment of BD. The further pharmaceutical composition may be a medicament which the patient may already be prescribed (e.g., an existing standard of care medication), and may itself be a composition comprising one or more active ingredients selected from those defined above. [0104] A compound of Formula (I) or a pharmaceutically acceptable salt thereof is synthesized according to the methods set forth in U.S. Patent No. 9,522,894, the disclosure of which is incorporated by reference in its entirety for all purposes. [0105] In various synthesis methods, functional groups can be protected and subsequently deprotected. The protection and deprotection of functional groups is described in ‘Protective Groups in Organic Synthesis’, 4 th Ed, T.W. Greene and P.G.M. Wuts, Wiley (2006) and ‘Protecting Groups’, 3 rd Ed P.J. Kocienski, Georg Thieme Verlag (2005), incorporated by reference herein in its entirety for all purposes. [0106] As provided throughout, according to the methods provided herein, a compound of Formula (I) can be administered as a pharmaceutically acceptable salt. A pharmaceutically acceptable salt of a compound of Formula (I) may be advantageous due to one or more of its chemical or physical properties, such as stability in differing temperatures and humidities, or a desirable solubility in H2O, oil, or other solvent. In some instances, a salt may be used to aid in the isolation or purification of the compound of Formula (I). [0107] Where the compound of Formula (I) is sufficiently acidic, pharmaceutically acceptable salts include, but are not limited to, an alkali metal salt, e.g., Na or K, an alkali earth metal salt, e.g., Ca or Mg, or an organic amine salt. Where the compound of Formula (I) is sufficiently basic, pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid addition salts. [0108] There may be more than one cation or anion depending on the number of charged functions and the valency of the cations or anions. [0109] For reviews on suitable salts, and pharmaceutically acceptable salts amenable for use herein, see Berge et al., J. Pharm. Sci., 1977, 66, 1-19 or “Handbook of Pharmaceutical Salts: Properties, selection and use”, P.H. Stahl, P.G. Vermuth, IUPAC, Wiley-VCH, 2002, incorporated by reference herein in its entirety for all purposes. [0110] The compounds of Formula (I) may form mixtures of its salt and co-crystal forms. It is also to be understood that the methods provided herein can employ such salt/co-crystal mixtures of the compound of Formula (I). [0111] Salts and co-crystals may be characterized using well known techniques, for example X-ray powder diffraction, single crystal X-ray diffraction (for example to evaluate proton position, bond lengths or bond angles), solid state NMR, (to evaluate for example, C, N or P chemical shifts) or spectroscopic techniques (to measure for example, O-H, N-H or COOH signals and IR peak shifts resulting from hydrogen bonding). [0112] It is also to be understood that certain compounds of Formula (I) may exist in solvated form, e.g., hydrates, including solvates of a pharmaceutically acceptable salt of a compound of Formula (I). [0113] In one embodiment, certain compounds of Formula (I) may exist as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. It is to be understood that the present disclosure encompasses all such isomeric forms. Certain compounds of Formula (I) may also contain linkages (e.g., carbon-carbon bonds, carbon- nitrogen bonds such as amide bonds) wherein bond rotation is restricted about that particular linkage, e.g., restriction resulting from the presence of a ring bond or double bond. Accordingly, it is to be understood that the methods provided herein can employ such isomers. Certain compound of Formula (I) may also contain multiple tautomeric forms. It is to be understood that the present disclosure encompasses all such tautomeric forms. Stereoisomers may be separated using conventional techniques, e.g., chromatography or fractional crystallization, or the stereoisomers may be made by stereoselective synthesis. [0114] In a further embodiment, the compounds of Formula (I) encompass any isotopically-labeled (or “radio-labelled”) derivatives of a compound of Formula (I). Such a derivative is a derivative of a compound of Formula (I) wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature. Examples of radionuclides that may be incorporated include 2H (also written as “D” for deuterium). As such, in one embodiment, a compound of Formula (I) is provided where one or more hydrogen atoms are replaced by one or more deuterium atoms; and the deuterated compound is used in one of the methods provided herein for treating BD. [0115] In a further embodiment, the compounds of Formula (I) may be administered in the form of a prodrug which is broken down in the human or animal body to give rise to a compound of the Formula (I). Examples of prodrugs include in vivo hydrolysable esters of a compound of the Formula (I). [0116] An in vivo hydrolysable (or cleavable) ester of a compound of Formula (I) that contains a carboxy or a hydroxy group is, for example, a pharmaceutically acceptable ester which is hydrolyzed in the human or animal body to produce the parent acid or alcohol. For examples of ester prodrugs derivatives, see: Curr. Drug. Metab. 2003, 4, 461, incorporated by reference herein in its entirety for all purposes. [0117] Various other forms of prodrugs are known in the art, and can be used in the methods provided herein. For examples of prodrug derivatives, see: Nature Reviews Drug Discovery 2008, 7, 255, the disclosure of which is incorporated by reference herein in its entirety for all purposes. EXAMPLES [0118] The present invention is further illustrated by reference to the following Examples. However, it should be noted that these Examples, like the embodiments described above, are illustrative and are not to be construed as restricting the scope of the invention in any way. Example 1 – Brensocatib Reduces Neutrophil Serine Protease Activities in Cultured Human Neutrophils [0119] To evaluate the pharmacological effects of brensocatib on surface PR3 expression and NSP activity, stem cells were differentiated to neutrophils in the presence of brensocatib. Primary bone marrow- or umbilical cord blood-derived CD34+ neutrophil progenitor cells (AllCells; Alameda, CA) were cultured for 7 days in STEMSPAN SFEM media (Stemcell Technologies; Tukwila, WA) supplemented with recombinant human Stem Cell Factor (Peprotech; Rocky Hill, NJ) and recombinant human IL-3 (Peprotech). The cells were differentiated in culture for another 7 days in STEMSPAN SFEM media (Stemcell Technologies) with recombinant human Granulocyte Colony Stimulating Factor (Peprotech), plus increasing concentrations of brensocatib (0 – ^^α ^0ααα α $Wα WKHα HQGα RIα WKHα differentiation/treatment period, cells were split into two aliquots for analysis. [0120] The first aliquot of differentiated neutrophils was harvested for flow cytometry to quantify PR3 expression on the surface of neutrophils. After harvest, the cells were resuspended in FACS staining buffer and Fc receptors were blocked for 30 min at 4 °C. Cells were stained for 30 min. with a mouse anti-PR3 monoclonal antibody (Abcam; Cambridge, MA) or a mouse monoclonal IgG isotype control (Abcam), followed by a Cy5-conjugated goat anti-mouse IgG secondary antibody (Abcam), and then analyzed using a Guava easyCyte 6HT flow cytometer (Millipore; Billerica, MA). [0121] For the second aliquot, neutrophils were lysed with 10% (v/v) Triton X-100 in PBS and the lysates were added to 96-well plates. Peptide substrates for NE (N-Methoxysuccinyl-Ala- Ala-Pro-Val-7-amido-4-methylcoumarin; Sigma-Aldrich; St. Louis, MO), for PR3 (aminobenzoyl-Val-Ala-Asp-Cys-Ala-Asp-Gln-Lys(2,4-dinitrophenyl); GenScript; Piscataway, NJ), and for CatG (N-Succinyl-Ala-Ala-Pro-Phe p-nitroanilide; Sigma) were added, and either fluorescence or absorbance was quantified using a Synergy microplate reader (BioTek; Winooski, VT). NSP inhibitors were used to test the specificity of enzymatic assays, including elastase inhibitor (Abcam) for NE, sivelestat (Abcam) for PR3, and cathepsin G inhibitor I (Cayman Chemical; Ann Arbor, MI) for CatG. A portion of cell lysate samples were retained for protein quantification using a Pierce™ BCA Protein Assay Kit (Thermo Fisher) according to the manufacturer’s instructions. The NSP activities were normalized to cell lysate protein concentrations and the final data were fit with nonlinear, four-parameter, logistic regression analysis by using GraphPad Prism software (GraphPad Software Inc.; La Jolla, CA). [0122] In a separate experiment, neutrophils were harvested, stained for 30 min with either a mouse anti-PR3 monoclonal antibody (Abcam; Cambridge, MA) or a mouse monoclonal IgG isotype control (Abcam), followed by a Cy5-conjugated goat anti-mouse IgG secondary antibody (Abcam), fixed in 4% formaldehyde, and mounted to glass slides with ProLong™ Diamond Antifade Mountant with DAPI (Thermo Fisher; Waltham, MA). Samples were imaged with an Axio fluorescence microscope (Zeiss; Oberkochen, Germany) using constant settings while imaging samples from different treatment conditions. [0123] The results of the fluorescence microscopy experiments demonstrated that neutrophils derived from either bone marrow or umbilical cord stem cells exhibited robust surface expression of PR3. Differentiation in the presence of 10 μM brensocatib markedly reduced surface PR3 in both cell types. [0124] Flow cytometry was employed to quantify mean fluorescence intensity (MFI) associated with surface PR3 expression in cells differentiated in the presence of brensocatib at various concentrations. Following normalization as percent of untreated cells, brensocatib concentration-dependently reduced surface PR3 expression in neutrophils derived from both bone marrow and umbilical cord blood stem cells, with maximum reductions greater than 60% (Figure 1). In addition, flow cytometry was used to evaluate the percentage of cells without surface PR3 expression. It was found that approximately 30-40% of untreated neutrophils expressed no detectable PR3 on their surface (Figure 2). Brensocatib concentration- dependently increased the percentage of cells with no detectable surface PR3 expression to more than 50% of neutrophils derived from cord blood stem cells and nearly 80% of neutrophils derived from bone marrow stem cells (Figure 2). [0125] In addition to measuring surface PR3, brensocatib effects on neutrophil serine protease (NSP) activities within the differentiated neutrophils was evaluated. Brensocatib concentration-dependently decreased the activities of NE and PR3 > 90%, and > 90%, respectively, at the highest concentration tested (10 μM; Figures 3-4). Example 2 – Therapeutic effects of brensocatib in a HSV model of BD [0126] In this example, a herpes simplex virus (HSV)-induced BD ICR mouse model was used to investigate the therapeutic effects of brensocatib. Specifically, ICR mice were infected with HSV to induce ulceration of mouth, genital, and skin, eye involvement, and other disease symptoms of BD in the mice, followed by treatment with brensocatib at two different doses (i.e., 10 mg brensocatib/kg body weight per mouse or 30 mg brensocatib/kg body weight per mouse) or a vehicle control. BD symptom severity and mortality were compared between the brensocatib-treated groups and the vehicle-treated control group. Materials and methods [0127] ICR mice were infected with HSV as described previously (Shim et al., Cytotherapy 13:835-847 (2011); Sohn et al., Eur J Dermatol.8(1):21-3 (1998); each of which is incorporated by reference herein in its entirety for all purposes). Briefly, earlobes of the mice were scratched with a needle, then inoculated with ^^α^/αof 1.0 x 106 plaque-forming units (pfu)/mL of HSV type l (F strain) solution. Ten days later, a second inoculation was performed using the same method. For virus inoculation, the mice were anesthetized by intraperitoneal injection with 2,2,2-tribromo ethanol (240 mg/kg). After viral inoculation, hydrocortisone (150 μg/mouse) was intraperitoneally injected once a day for 5 days. The mice were kept in a conventional temperature- and light-controlled room (20-22 °C, 12 hr. light cycle starting at 8:00 a.m.). The mice had free access to food and water, and were handled in accordance with a protocol approved by an institutional animal care committee. [0128] Symptoms of BD were confirmed by follow-up for 5 to 16 weeks after the second virus inoculation. Several symptoms, including oral ulcers, genital ulcers, erythema, skin pustules, skin ulcers, arthritis, diarrhea, red-eye, loss of balance, and facial edema, were observed in the mice after HSV inoculation. Oral, genital, skin ulcers, and eye symptoms were classified as major symptoms. Arthritis, intestinal ulceration, and neurological involvement were considered as minor symptoms. Mice with at least one major symptom and at least one minor symptom were classified as having BD. Each symptom was scored as one point. The sum of points was used to determine the severity of BD using 2006 BD Current Activity Form prepared by the International Society for Behçet’s Disease
Figure imgf000024_0001
The disappearance of symptoms or a reduction in skin lesion size of 20% or more indicated improvement of BD. [0129] Brensocatib was dissolved in a tartaric acid solution. When BD symptoms appeared in the mice, the mice were administered the brensocatib solution at 10 mg brensocatib/kg body weight per mouse or 30 mg brensocatib/kg body weight per mouse, or a vehicle solution containing the same concentration of tartaric acid (control group). There were 11, 11, and 15 BD mice in the 10 mg brensocatib, 30 mg brensocatib, and vehicle-treated groups, respectively, at the start of the administration period. Dosing lasted for 4 weeks, with the brensocatib solution or the vehicle solution administered orally twice a day for 2 weeks from the start of the administration, and once a day for the next 2 weeks. Monitoring of BD symptoms was continued throughout the administration period to determine whether any improvement(s) occurred. Mortality rates among the study groups were also determined. After 4 weeks of administration, the mice were sacrificed, and tissue samples, including bone marrows, plasma, and blood cells, were collected from the mice for analysis. Results 1. Brensocatib treatment reduced mortality in HSV-induced BD mice [0130] Figure 5 shows the percentage of surviving BD mice during the treatment with the vehicle control (dotted line), 10 mg/kg brensocatib (thick solid line), or 30 mg/kg brensocatib (thin solid line). Brensocatib treatment at both doses resulted in lower mortality rates (0% in the 10 mg/kg group and 18.2% in the 30 mg/kg group) than the vehicle treatment (33.3% mortality rate) at the end of the treatment period. More notably, mice treated with 10 mg/kg brensocatib displayed 0% mortality rate throughout the treatment period, which was significantly lower as compared to the vehicle control group, with P= 0.0481 using LogRank (Mantel-Cox) test. 2. Brensocatib treatment resulted in higher percentages of HSV-induced BD mice with improvement and/or no change in symptoms [0131] Table 1 shows that, as compared to the vehicle control, brensocatib treatment at both doses resulted in higher percentages of HSV-induced BD mice with improvement in BD symptoms, no change in BD symptoms, and/or a combination of the two, at 2 weeks and 4 weeks of the administration period, based on the disease severity scores. Moreover, brensocatib treatment at the 10 mg/kg dose was more effective than at the 30 mg/kg dose in improving the BD symptoms, consistent with their effects on the reduction in mortality. Table 1. Efficacy of brensocatib on symptoms in HSV-induced BD mice based on disease severity score
Figure imgf000025_0001
Example 3 – Prophylactic effects of brensocatib in a HSV model of BD [0132] In this example, a similar HSV-induced BD C57BL/6 mouse model is used to investigate the prophylactic effects of brensocatib. HSV-induced BD occurrence rate in C57BL/6 mice is estimated to be about 40%, as compared to a 10-15% occurrence rate in ICR mice used in Example 2. [0133] Specifically, C57BL/6 mice in groups of about 30 each are treated with brensocatib at three different doses, i.e., 3 mg/kg, 10 mg/kg, and 30 mg/kg per day, or a vehicle control solution containing tartaric acid (as described in Example 2), via oral administration twice per day, seven days prior to a first inoculation of HSV performed as described in Example 2. Ten days later a second HSV inoculation is performed also as described in Example 2. After the first inoculation of HSV, brensocatib or vehicle treatment is performed 2 times per day for 7 days, and thereafter once a day for the next 16 weeks. Following the second HSV inoculation, BD occurrence rates are determined, BD symptoms are monitored, the disease severity is scored (as described in Example 2), and mortality rates are recorded among the study groups for the next 16 weeks. Thereafter, the mice are sacrificed, and tissue samples, including bone marrows, plasma, and blood cells, are collected from the mice for analysis. * * * * * * * [0134] All, documents, patents, patent applications, publications, product descriptions, and protocols which are cited throughout this application are incorporated herein by reference in their entireties for all purposes. [0135] The embodiments illustrated and discussed in this specification are intended only to teach those skilled in the art the best way known to the inventors to make and use the invention. Modifications and variation of the above-described embodiments of the invention are possible without departing from the invention, as appreciated by those skilled in the art in light of the above teachings. It is therefore understood that, within the scope of the claims and their equivalents, the invention may be practiced otherwise than as specifically described.

Claims

CLAIMS 1. A method for treating Behçet’s Disease (BD) in a patient in need of treatment, comprising, administering to the patient a pharmaceutical composition comprising an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof,
Figure imgf000027_0001
R2 is hydrogen, F, Cl, Br, OSO2C1-3alkyl, or C1-3alkyl; R3 is hydrogen, F, Cl, Br, CN, CF3, SO2C1-3alkyl, CONH2 or SO2NR4R5, wherein R4 and R5 together with the nitrogen atom to which they are attached form an azetidine, pyrrolidine or piperidine ring; or R6 is C1-3alkyl, optionally substituted by 1, 2 or 3 F and/or optionally by OH, OC1-3alkyl, N(C1- 3alkyl)2, cyclopropyl, or tetrahydropyran; R7 is hydrogen, F, Cl or CH3; X is O, S or CF2; Y is O or S; and Q is CH or N.
Figure imgf000028_0002
Q ; R6 is C1-3alkyl, wherein the C1-3alkyl is optionally substituted by 1,
2 or 3 F and optionally by one substituent selected from OH, OC1-3alkyl, N(C1-3alkyl)2, cyclopropyl, or tetrahydropyran; and R7 is hydrogen, F, Cl or CH3.
3. The method of claim 1 or 2, wherein,
Figure imgf000028_0001
X is O, S or CF2; Y is O or S; R6 is C1-3alkyl, wherein the C1-3alkyl is optionally substituted by 1, 2 or 3 F and optionally by one substituent selected from OH, OC1-3alkyl, N(C1-3alkyl)2, cyclopropyl, or tetrahydropyran; and R7 is hydrogen, F, Cl or CH3.
4. The method of any one of claims 1-3, wherein,
Figure imgf000028_0003
5. The method of any one of claims 1-4, wherein X is O, S or CF2; R6 is C1-3alkyl, wherein the C1-3alkyl is optionally substituted by 1, 2 or 3 F; and R7 is hydrogen, F, Cl or CH3.
6. The method of any one of claims 1-4, wherein X is O; R6 is C1-3alkyl, wherein the C1- 3alkyl is optionally substituted by 1, 2 or 3 F; and R7 is hydrogen.
7. The method of any one of claims 1-4, wherein X is O; R6 is C1-3alkyl; and R7 is hydrogen.
8 The method of claim 1, wherein the compound of Formula (I) is selected from the group consisting of (2S)-N-[(1S)-1-Cyano-2-(4’-cyanobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl}- 1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(3,7-dimethyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; 4’-[(2S)-2-Cyano-2-{[(2S)-1,4-oxazepan-2-ylcarbonyl]amino}ethyl]biphenyl-3-yl methanesulfonate; (2S)-N-{(1S)-1-Cyano-2-[4-(3-methyl-1,2-benzoxazol-5-yl)phenyl]ethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4’-(trifluoromethyl)biphenyl-4-yl]ethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-[(1S)-1-Cyano-2-(3’,4’-difluorobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(6-cyanopyridin-3-yl)phenyl]ethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(4-methyl-3-oxo-3,4-dihydro-2H-1,4-benzothiazin-6- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(3-ethyl-7-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2-hydroxy-2-methylpropyl)-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2,2-difluoroethyl)-7-fluoro-2-oxo-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-(4-{3-[2-(dimethylamino)ethyl]-2-oxo-2,3-dihydro-1,3-benzoxazol- 5-yl}phenyl)ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(3,3-difluoro-1-methyl-2-oxo-2,3-dihydro-1H-indol-6- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(7-fluoro-3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(3-ethyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl}- 1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[3-(cyclopropylmethyl)-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2-methoxyethyl)-2-oxo-2,3-dihydro-1,3-benzothiazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[2-oxo-3-(propan-2-yl)-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(4-methyl-3-oxo-3,4-dihydro-2H-1,4-benzoxazin-6- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2-methoxyethyl)-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(5-cyanothiophen-2-yl)phenyl]ethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-[(1S)-2-(4’-Carbamoyl-3’-fluorobiphenyl-4-yl)-1-cyanoethyl]-1,4-oxazepane-2- carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(1-methyl-2-oxo-1,2-dihydroquinolin-7-yl)phenyl]ethyl}-1,4- oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[2-oxo-3-(tetrahydro-2H-pyran-4-ylmethyl)-2,3-dihydro-1,3- benzoxazol-5-yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-2-[4-(7-Chloro-3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]-1- cyanoethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[3-(2,2-difluoroethyl)-2-oxo-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-[(1S)-1-Cyano-2-{4-[2-oxo-3-(2,2,2-trifluoroethyl)-2,3-dihydro-1,3-benzoxazol-5- yl]phenyl}ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzothiazol-5- yl)phenyl]ethyl}-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-1-Cyano-2-[4’-(methylsulfonyl)biphenyl-4-yl]ethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-{(1S)-2-[4’-(Azetidin-1-ylsulfonyl)biphenyl-4-yl]-1-cyanoethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-[(1S)-1-Cyano-2-(4’-fluorobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide; (2S)-N-{(1S)-2-[4-(1,3-Benzothiazol-5-yl)phenyl]-1-cyanoethyl}-1,4-oxazepane-2- carboxamide; (2S)-N-[(1S)-1-Cyano-2-(4’-cyanobiphenyl-4-yl)ethyl]-1,4-oxazepane-2-carboxamide; and pharmaceutically acceptable salts thereof.
9. The method of claim 1, wherein the compound of Formula (I) is brensocatib; or a pharmaceutically acceptable salt thereof.
10. The method of claim 1, wherein the compound of Formula (I) is brensocatib .
11. The method of any one of claims 1-10, wherein the composition comprises a pharmaceutically acceptable adjuvant, diluent or carrier.
12. The method of any one of claims 1-11, wherein the administering comprises oral administration.
13. The method of any one of claims 1-12, wherein the administering to the patient is carried out one time daily.
14. The method of any one of claims 1-12, wherein the administering to the patient is carried out two times daily.
15. The method of any one of claims 1-12, wherein the administering to the patient is carried out once, every other day.
16. The method of any one of claims 1-12, wherein the administering to the patient is carried out once every third day.
17. The method of any one of claims 1-16, wherein the administering is carried out for about 3 months.
18. The method of any one of claims 1-16, wherein the administering is carried out for about 6 months.
19. The method of any one of claims 1-16, wherein the administering is carried out for about 9 months.
20. The method of any one of claims 1-16, wherein the administering is carried out for about 12 months.
21. The method of any one of claims 1-16, wherein the administering is carried out for about 18 months.
22. The method of any one of claims 1-16, wherein the administering is carried out for about 24 months.
23. The method of any one of claims 1-22, further comprising administering one or more additional active agents to the patient in need of treatment.
24. The method of claim 23, wherein the one or more additional active agents comprises colchicine, levamisole hydrochloride, a corticosteroid, chlorambucil, cyclophosphamide, cyclosporine, azathioprine or thalidomide.
25. The method of claim 23, wherein the one or more additional active agents comprises an NSAID.
26. The method of claim 23, wherein the one or more additional active agents comprises an immunosuppressive agent.
27. The method of claim 26, wherein the immunosuppressive agent is a 71)ĮαLQKLELWRUααα 28. The method of claim 27, wherein the 71)ĮαLQKLELWRUαLVαLQIOL[LPDE. 29. The method of claim 27, wherein the 71)ĮαLQKLELWRUαLVαadalimumab. 30. The method of claim 27, wherein the 71)ĮαLQKLELWRUαLVαetanercept. 31. The method of claim 26, wherein the immunosuppressive agent is thalidomide. 32. The method of claim 26, wherein the immunosuppressive agent is azathioprine. 33. The method of any one of claims 23-32, wherein the one or more additional active agents comprises colchicine. 34. The method of any one of claims 23-33, wherein the one or more additional active agents comprises interferon alfa-2a.
PCT/US2021/064810 2020-12-22 2021-12-22 Certain (25)-iv-[(ls')~ i-c yan0-2-phenylethyl·]- 1,4-oxazepane-2- carboxamides for treating behcet's disease WO2022140516A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063129133P 2020-12-22 2020-12-22
US63/129,133 2020-12-22

Publications (1)

Publication Number Publication Date
WO2022140516A1 true WO2022140516A1 (en) 2022-06-30

Family

ID=82158400

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/064810 WO2022140516A1 (en) 2020-12-22 2021-12-22 Certain (25)-iv-[(ls')~ i-c yan0-2-phenylethyl·]- 1,4-oxazepane-2- carboxamides for treating behcet's disease

Country Status (1)

Country Link
WO (1) WO2022140516A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11655221B2 (en) 2014-01-24 2023-05-23 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150210655A1 (en) * 2014-01-24 2015-07-30 Astrazeneca Ab Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
WO2020018551A1 (en) * 2018-07-17 2020-01-23 Insmed Incorporated Certain (2s)- n-[(1s)-1-cyano-2-phenylethyl]-1,4- oxazepane-2-carboxamides for treating inflammatory bowel disease

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150210655A1 (en) * 2014-01-24 2015-07-30 Astrazeneca Ab Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
WO2020018551A1 (en) * 2018-07-17 2020-01-23 Insmed Incorporated Certain (2s)- n-[(1s)-1-cyano-2-phenylethyl]-1,4- oxazepane-2-carboxamides for treating inflammatory bowel disease

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
AVCI DENIZ: "Dipeptidyl Peptidase-4 Inhibitors and Inflammation: Dpp-4 Inhibitors Improve Mean Pleatelet Volume and Gamma Glutamyl Transferase Level", JOURNAL OF BIOSCIENCES AND MEDICINES, vol. 07, no. 02, 1 February 2019 (2019-02-01), pages 42 - 53, XP055954746, ISSN: 2327-5081, DOI: 10.4236/jbm.2019.72004 *
E. ALPSOY: "Behçet’s disease: Treatment of mucocutaneous lesions", CLINICAL AND EXPERIMENTAL RHEUMATOLOGY, vol. 23, no. 4, 1 July 2005 (2005-07-01), pages 532 - 539, XP055954742 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11655221B2 (en) 2014-01-24 2023-05-23 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11655222B2 (en) 2014-01-24 2023-05-23 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11655223B2 (en) 2014-01-24 2023-05-23 Astrazeneca Ab Certain (2S)-N-[(1 s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11655224B2 (en) 2014-01-24 2023-05-23 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11667615B2 (en) 2014-01-24 2023-06-06 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11673871B2 (en) 2014-01-24 2023-06-13 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11673872B2 (en) 2014-01-24 2023-06-13 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11680049B2 (en) 2014-01-24 2023-06-20 Astrazeneca Ab Certain (2S)-N-[(1S)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11773069B2 (en) 2014-01-24 2023-10-03 Astrazeneca Ab Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors
US11814359B2 (en) 2014-01-24 2023-11-14 Astrazeneca Ab Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides as dipeptidyl peptidase 1 inhibitors

Similar Documents

Publication Publication Date Title
US20210186984A1 (en) Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides for treating anca associated vasculitides
US9233089B2 (en) Treatment of pulmonary hypertension with leukotriene inhibitors
KR20150046039A (en) Nep inhibitors for treating diseases characterized by atrial enlargement or remodeling
KR20050042154A (en) Synergistic combination of an alpha-2-delta ligand and a pdev inhibitor for use in the treatment of pain
US20210252015A1 (en) Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides for treating inflammatory bowel disease
KR20190035781A (en) (2S) -N- [(1S) -1-cyano-2-phenylethyl] -1,4-oxazepane-2-carboxamide
JP7336450B2 (en) (2S)-N-{(1S)-1-cyano-2-[4-(3-methyl-2-oxo-2,3-dihydro-1,3-benzoxazol-5-yl)phenyl]ethyl} -Pharmaceutical composition containing 1,4-oxazepane-2-carboxamide
TW201235040A (en) Drug therapy for preventing or treating glaucoma
US20210322438A1 (en) Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides for treating lupus nephritis
WO2022140516A1 (en) Certain (25)-iv-[(ls&#39;)~ i-c yan0-2-phenylethyl·]- 1,4-oxazepane-2- carboxamides for treating behcet&#39;s disease
JP2023540393A (en) Method for treating metastases with cathepsin C inhibitors
TW200946113A (en) PAI-1 expression and activity inhibitors for the treatment of ocular disorders
CA3215375A1 (en) Certain n-(1-cyano-2-phenylethyl)-1,4-oxazepane-2-carboxamides for treating cystic fibrosis
JP4966019B2 (en) Preventive and therapeutic agents for dry eye in chronic graft-versus-host disease
WO1998051313A1 (en) Remedies for dry eye
WO2023159120A1 (en) Certain n-(1-cyano-2-phenylethyl)-1,4-oxazepane-2-carboxamides for treating hidradenitis suppurativa
WO2023076615A1 (en) Certain n-(1-cyano-2-phenylethyl)-1,4-oxazepane-2-carboxamides for treating chronic rhinosinusitis
CA3236069A1 (en) Certain n-(1-cyano-2-phenylethyl)-1,4-oxazepane-2-carboxamides for treating chronic rhinosinusitis
EP4182700A1 (en) Methods for extracting neutrophil serine proteases and treating dipeptidyl peptidase 1-mediated conditions

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: 21912127

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: 21912127

Country of ref document: EP

Kind code of ref document: A1