WO2021076124A1 - Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp) - Google Patents

Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp) Download PDF

Info

Publication number
WO2021076124A1
WO2021076124A1 PCT/US2019/056533 US2019056533W WO2021076124A1 WO 2021076124 A1 WO2021076124 A1 WO 2021076124A1 US 2019056533 W US2019056533 W US 2019056533W WO 2021076124 A1 WO2021076124 A1 WO 2021076124A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrimidin
pyrazol
pyrrolo
jak1
pharmaceutically acceptable
Prior art date
Application number
PCT/US2019/056533
Other languages
French (fr)
Inventor
Paul Smith
Joerg Wenzel
Original Assignee
Incyte Corporation
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 Incyte Corporation filed Critical Incyte Corporation
Priority to JP2022523053A priority Critical patent/JP2023506118A/en
Priority to PCT/US2019/056533 priority patent/WO2021076124A1/en
Publication of WO2021076124A1 publication Critical patent/WO2021076124A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • the present application provides methods for the treatment of cutaneous lupus erythematosus and/or Lichen planus (LP) using compounds that modulate the activity of Janus kinase (JAK) 1.
  • Cutaneous lupus erythematosus is an inflammatory autoimmune skin disease with heterogenic subtypes varying from localized discoid plaques to severe and widespread erythrosquamous lesions in affected patients (see e.g, Kuhn & Landmann, J. Autoimmun. 2014, 48-49:14-19).
  • the disease is characterized by a typical histological pattern called “interface dermatitis” (ID) consisting of colloid bodies and an anti-epidermal cytotoxic lymphocytic infiltrate in the dermoepidermal junction which is orchestrated by IFN-regulated proinflammatory cytokines (see e.g. , Wenzel & Tilting, J. Invest. Dermatol. 2008, 128:2392-2402; and Wenzel et al, Br. J. Dermatol. 2007, 157:752-757).
  • ID interface dermatitis
  • CLE is of multifactorial origin; including both genetic and environmental risk factors (see e.g., Kunz et al, Exp. Dermatol. 2015, 24:510-515; Sinha & Dey-Rao, J. Investig. Dermatol. Symp. Proc., 2017, 18:S75-S80; and Szczqch et al, Lupus , 2017, 26:791-807).
  • UV radiation promotes cellular damage, apoptosis and release of DNA-containing blebs. It is considered that cell debris clearance is impaired entailing secondary necrosis (see e.g., Kuhn et al, Arthritis Rheum.
  • keratinocytes and particularly plasmacytoid dendritic cells react on DAMPs inappropriately with immense type I IFN production through TLR-dependent or TLR-independent pathways in CLE (see e.g., Mustelin et al, Front. Immunol. 2019, 10:238; Wollenberg et al, J. Invest. Dermatol. 2002, 119: 1096-1102; Wenzel et al, Arch. Dermatol. Res. 2009, 301:83-86; Yu et al, J. Autoimmun. 2013, 41:34-45; and Katayama et al, J. Invest. Dermatol. DOI: 10.1016/j jid.2019.02.035).
  • ACR American College of Rheumatology
  • SLE Systemic Lupus Erythematosus
  • the ACR guidelines require four of eleven criteria to be met for a diagnosis of SLE, however, only four of the criteria are cutaneous in nature (malar rash, discoid lesions, mucosal ulcers, and photosensitivity) and therefore involvement of skin pathologies is not obligatory for the diagnosis of SLE.
  • CLE cutaneous lupus erythematosus subtypes can occur in the absence of systemic disease (SLE); CLE is two to three times more frequent than SLE (see e.g ., Tebbe & Orfanos, Lupus , 1997, 6(2):96-104).
  • the present application provides methods of treating a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a JAK1 selective inhibitor provided herein.
  • a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a JAK1 selective inhibitor provided herein.
  • FIGs. 1 A-1D show the functional role of JAK1 expression in CLE skin lesions.
  • FIG. 1C shows upregulated expression of IFN-associated genes in CLE lesional skin (2-fold, / ⁇ 0,01 ; Welch’s /-test) compared to healthy controls (HC).
  • FIG. ID shows KEGG pathways upregulated in CLE. KEGG pathway classification was performed using Database for Annotation, Visualization and Integrated Discovery (DAVID ver. 6.8). C-values were generated with EASE Score. Count: number of genes >2-fold upregulated in CLE within the respective KEGG pathway.
  • FIGs. 2A-2E show the effects of pharmacological JAK1 inhibition on proinflammatory cytokine expression and IFN-associated gene regulation in cultured keratinocytes.
  • FIG. 2A shows mean area intensity in pixel values of anti-pJAKl- antibody staining in stimulated HaCaT (eNA), stimulated and inhibitor-treated HaCaT (eNA+ JAKl inhibitor ⁇ l- ⁇ l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl ⁇ -3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl ⁇ acetonitrile (i.e., INCB039110); eNA+ JAKl/2, inhibitor: ruxolitinib) and unstimulated HaCaT (C-Medium). Measurement was performed out using Fiji software
  • FIG. 2B shows representative immunofluorescence micrographs of eNA-stimulated untreated (eNA) or inhibitor-treated (eNA+JAKl, inhibitor:INCB039110; eNA+JAKl/2, inhibitonruxolitinib) HaCaT as well as unstimulated HaCaT-cells (Medium Control).
  • FIG. 2C shows expression of CLE-typical genes within eNA-stimulated NHEK (+, 4 samples) and >2-fold downregulated expression (p ⁇ 0,05; Welch's /-test) after treatment of stimulated NHEK with JAKl selective INCB039110 (JAKl; 4 samples).
  • FIG. 2D shows the effect of different JAK inhibitors on CXCL10 expression in stimulated HaCaT after 24 hours of incubation. Cells were treated with JAKl selective INCB039110 (JAKl), JAKl/2 selective ruxolitinib (JAK 1/2) and JAK3 selective FM- 381 (JAK3). Measured by ELISA.
  • FIGs. 3A-3C show pharmacological JAKl inhibition in vivo in a lupus-prone TREX1 mouse model.
  • FIG. 3B shows mean score of the mouse
  • the present application provides, inter alia , methods of treating a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering a therapeutically effective amount of JAK1 selective inhibitor, or a pharmaceutically acceptable salt thereof.
  • the disease is cutaneous lupus erythematosus (CLE).
  • the cutaneous lupus erythematosus (CLE) is selected from acute cutaneous lupus erythematosus, subacute cutaneous lupus erythematosus, and chronic cutaneous lupus erythematosus.
  • the cutaneous lupus erythematosus is acute cutaneous lupus erythematosus.
  • the cutaneous lupus erythematosus is selected from subacute cutaneous lupus erythematosus (SCLE) and chronic discoid lupus erythematosus (CDLE).
  • the cutaneous lupus erythematosus is subacute cutaneous lupus erythematosus (SCLE).
  • the cutaneous lupus erythematosus is chronic discoid lupus erythematosus (CDLE).
  • the disease is Lichen planus (LP).
  • cutaneous lupus refers to a form of the disease in which symptoms are restricted to those that affect the skin.
  • a patient may be diagnosed with cutaneous lupus, but that does not mean that he or she has SLE, which affects multiple parts of the body. Similarly, if a patient has SLE, it does not mean that he or she will necessarily have cutaneous lupus.
  • CLASI Cutaneous Lupus Area and Severity Index
  • Cutaneous lupus is divided into several subtypes, including acute cutaneous lupus erythematosus, subacute cutaneous lupus erythematosus, and chronic cutaneous lupus erythematosus.
  • Acute cutaneous LE typically presents in the third decade of life and is frequently associated with active SLE.
  • the most typical form of acute cutaneous lupus is a malar rash-flattened areas of red skin on the face that resemble a sunburn (see e.g. , Fabbri et al., Am. ./. Clin. Dermatol. 2003, 4(7):449-65). These lesions are typically transient, sun-induced, and non-scarring.
  • Chronic cutaneous lupus includes discoid LE (DLE), LE profundus (LEP), chilblain LE (CELLE), and LE tumidus (LET).
  • the chronic cutaneous lupus is selected from discoid lupus erythematosus (DLE), lupus erythematosus profundus (LEP), chilblain lupus erythematosus (CHLE), and lupus erythematosus tumidus (LET).
  • Discoid LE lesions are the most common lesions of Chronic cutaneous lupus erythematosus (CCLE) and appear as disk-shaped, round lesions. DLE occurs more frequently in women in their fourth and fifth decade of life. Patients with DLE generally have a more benign disease course as compared to patients with other CLE subtypes, with only most patients (-90-95%) never develop lupus in other organ systems (SLE) symptoms (see e.g. , Crowson & Magro, ./. Cutan. Pathol. 2001,
  • LE profundus or panniculitis
  • LEP tends to have a chronic course, characterized by remission and flares, and ultimately leaving atrophic scars (see e.g. , Fabbri et al., Am. J. Clin. Dermatol. 2003, 4(7):449-65). Histology shows lobular panniculitis with a dense lymphocytic infiltrate.
  • Chilblain lupus is a rare form of CCLE resembling frostbite. Lesions appear as painful, violaceous plaques and nodules in cold-exposed areas. Central erosions or ulcerations may occur on acral surfaces, such as fingers, toes, heels, nose, and ears. Chilblain lupus occurs when there is a temperature drop, and can be difficult to distinguish from frostbite. Pathology shows epidermal atrophy, interface vacuolization, and a perivascular mononuclear infiltrate. Approximately 80% of patients with CHLE will not develop SLE (see e.g., Hedrich et al., Clin. Rheumatol. 2008, 27(8):949-54).
  • Lupus tumidus is a subtype of CCLE characterized by extreme photosensitivity and a benign course occurring preferentially in men, unlike SLE which primarily occurs in women. Clinically, these lesions appear on the face as erythematous, edematous, urticaria-like polycylic plaques with sharp raised borders and smooth surfaces.
  • SCLE subacute cutaneous lupus
  • the lesions are described as having a scaly red annular (“ring-like”) redness with central clearing.
  • the lesions may also be polycyclic - that is, having the appearance of multiple rings coming together.
  • annular and papulosquamous are two morphologic variants of SCLE: annular and papulosquamous.
  • the annular variant is characterized by scaly annular erythematous plaques, which tend to coalesce and produce a polycyclic array (see e.g., Walling & Sontheimer, Am. J. Clin. Dermatol. 2009, 10(6):365-81).
  • the papulosquamous variant can resemble eczema, as well as pityriasis in some instances (see e.g., Caproni et al. Int. J. Dermatol. 2001, 40(l):59-62).
  • SCLE lesions occur in sun-exposed areas, including the upper thorax (‘V’ distribution), upper back, and the extensor surfaces of arms and forearms. About 10% of SLE patients will have SCLE lesions and those patients with systemic disease tend to have only mild symptoms in other organ compartments.
  • Anti-IFNa-agents e.g, sifalimumab, rontalizumab
  • these agents reduced the IFN signature in the blood but had limited effect on disease activity. Without being bound by theory, this was possibly due to the high variability of the different type I IFNs, including not only IFNa but also IFNP and IFNK binding to the same receptors (see e.g., Kalunian et al, Ann. Rheum. Dis. 2016, 75:196-202; Merrill et al, Ann. Rheum. Dis. 2011, 70:1905-1913; and Sarkar et al, Ann.
  • the JAK family consists of four non-receptor tyrosine kinases (JAKl-3, TYK2) that transduce signals from growth factors and cytokines such as type I/III IFNs.
  • JAK inhibitors were initially developed for the treatment of haematologic diseases with definite JAK-mutations showing anti clonal activity (see e.g, Morgan et al, Annu. Rev. Med. 2008, 59:213-222; and Pardanani et al, Leukemia 2008, 22:23- 30), but they also provide significant immunosuppressive effects (see e.g, Santos & Verstovsek, Expert Opin. Pharmacother. 2014, 15:1465-1473; Schwartz et al, Nat. Rev.
  • JAK inhibitors provided beneficial effects in interface dermatitides including graft-versus- host disease (see e.g., Spoerl et al, Blood , 2014, 123:3832-3842), dermatomyositis (see e.g., Hornung et al, N. Engl. J. Med. 2014, 371:2537-2538; and Homung et al, N. Engl. J. Med. 2015, 372:1274) and LE (see e.g., Wenzel et al, J. Invest. Dermatol.
  • CLASI Cutaneous Lupus Erythematosus Disease Area and Severity Index
  • JAK1 inhibitors e.g ., JAKl selective inhibitors.
  • the compound is selected from:
  • the compounds or salt is selective for JAK1 over JAK2, JAK3 and TYK2.
  • JAK1 plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states.
  • IL-6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (see e.g ., Fonesca, et al., Autoimmunity Reviews, 8:538-42, 2009).
  • IL-6 signals, at least in part, through JAK1, IL-6 can be indirectly through JAK1 inhibition, resulting in potential clinical benefit (see e.g, Guschin, et al. Embo J 14:1421, 1995; and Smolen, et al. Lancet 371:987, 2008).
  • JAK1 inhibition In other autoimmune diseases and cancers, elevated systemic levels of inflammatory cytokines that activate JAK1 may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAK1 inhibition.
  • Selective inhibitors of JAK1 may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases, as described herein.
  • the compounds or salt inhibits JAK1 preferentially over JAK2 (e.g, having a JAK2/JAK1 IC50 ratio >1).
  • the compounds or salts provided herein are about 10-fold more selective for JAK1 over JAK2.
  • the compounds or salts provided herein are about 3- fold, about 5-fold, about 10-fold, about 15-fold, or about 20-fold more selective for JAK1 over JAK2 as calculated by measuring IC50 at 1 mM ATP (see Example 1).
  • the JAK1 inhibitor is a compound of Table 1, or a pharmaceutically acceptable salt thereof.
  • the compounds in Table 1 are JAK1 selective inhibitors (e.g, selective over JAK2, JAK3, and TYK2).
  • the IC50 values obtained by the method of Example 1 at 1 mM ATP are shown in Table 1.
  • the JAK1 inhibitor is ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the JAK1 inhibitor is (l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile adipic acid salt.
  • the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide, or a pharmaceutically acceptable salt thereof.
  • the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide phosphoric acid salt.
  • the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide hydrochloric acid salt.
  • the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide hydrobromic acid salt.
  • the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide sulfuric acid salt.
  • the JAK1 inhibitor is ((2R,5S)-5- ⁇ 2-[(lR)-l- hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl ⁇ tetrahydro-2H-pyran-2- yl)acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the JAK1 inhibitor is ((2R,5S)-5- ⁇ 2-[(lR)-l- hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl ⁇ tetrahydro-2H-pyran-2- yl)acetonitrile monohydrate.
  • the JAK1 inhibitor is selected from the compounds, or pharmaceutically acceptable salts thereof, of US Patent Publ. No. 2011/0224190, filed March 9, 2011, US Patent Publ. No. 2014/0343030, filed May 16, 2014, US Patent Publ. No. 2014/0121198, filed October 31, 2013, US Patent Publ. No. 2010/0298334, filed May 21, 2010, US Patent Publ. No. 2011/0059951, filed August 31, 2010, US Patent Publ. No. 2012/0149681, filed November 18, 2011, US Patent Publ. No. 2012/0149682, filed November 18, 2011, US Patent Publ. 2013/0018034, filed June 19, 2012, US Patent Publ. No. 2013/0045963, filed August 17, 2012, and US Patent Publ. No. 2014/0005166, filed May 17, 2013, each of which is incorporated herein by reference in its entirety.
  • the JAK1 inhibitor is a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein: X is N or CH;
  • A is phenyl, pyridinyl, or pyrimidinyl each of which is optionally substituted with 1 or 2 independently selected R 1 groups; and each R 1 is, independently, fluoro, or trifluoromethyl.
  • the compound of Formula I is ⁇ l- ⁇ l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl ⁇ -3[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl ⁇ acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the compound of Formula I is 4- ⁇ 3-(Cyanomethyl)-3- [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl ⁇ -N-[4-fluoro-2- (trifluoromethyl)phenyl]piperidine-l -carboxamide, or a pharmaceutically acceptable salt thereof.
  • the compound of Formula I is [3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]- 1 -( 1 - ⁇ [2-(trifluoromethyl)pyrimidin-4- yl]carbonyl ⁇ piperidin-4-yl)azetidin-3-yl]acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the JAK1 inhibitor is a compound of Formula II:
  • R 2 is Ci-6 alkyl, Ci-6 haloalkyl, C3- 6 cycloalkyl, or C3- 6 cycloalkyl-Ci- 3 alkyl, wherein said Ci-6 alkyl, C 3 - 6 cycloalkyl, and C 3 - 6 cycloalkyl-Ci- 3 alkyl, are each optionally substituted with 1, 2, or 3 substituents independently selected from fluoro, - CF 3 , and methyl;
  • R 3 is H or methyl;
  • R 4 is H, F, or Cl;
  • R 5 is H or F;
  • R 6 is H or F;
  • R 7 is H or F
  • R 8 is H or methyl
  • R 9 is H or methyl
  • R 10 is H or methyl; and R 11 is H or methyl.
  • the compound of Formula II is 4-[3-(cyanomethyl)-3- (3',5'-dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide , or a pharmaceutically acceptable salt thereof.
  • the JAK1 inhibitor is a compound of Formula III: or a pharmaceutically acceptable salt thereof, wherein:
  • Cy 4 is a tetrahydro-2H-pyran ring, which is optionally substituted with 1 or 2 groups independently selected from CN, OH, F, Cl, C1-3 alkyl, C1-3 haloalkyl, CN-C1- 3 alkyl, HO-C1- 3 alkyl, amino, C1- 3 alkylamino, and di(Ci-3 alkyl)amino, wherein said Ci-3 alkyl and di(Ci-3 alkyl)amino is optionally substituted with 1, 2, or 3 substituents independently selected from F, Cl, C1- 3 alkylaminosulfonyl, and C1- 3 alkyl sulfonyl; and
  • R 12 is -CH2-OH, -CH(CH 3 )-OH, or -CH2-NHSO2CH3.
  • the compound of Formula III is ((2R,5S)-5- ⁇ 2-[(lR)-l- hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl ⁇ tetrahydro-2H-pyran-2- yl)acetonitrile, or a pharmaceutically acceptable salt thereof.
  • the compound includes at least one deuterium atom. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1-2, 1-3, 1-4, 1-5, or 1-6 deuterium atoms. In some embodiments, all of the hydrogen atoms in a compound can be replaced or substituted by deuterium atoms.
  • substitution with heavier isotopes may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances (see e.g ., A. Kerekes et. al. J. Med. Chem. 2011, 54, 201-210; R. Xu et. al. J. Label Compd. Radiopharm. 2015, 58, 308-312).
  • substitution at one or more metabolism sites may afford one or more of the therapeutic advantages.
  • the JAK1 inhibitor (e.g. , the JAK1 selective inhibitor) is a compound, wherein one or more hydrogen atoms in the compound are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof.
  • the phrase “optionally substituted” means unsubstituted or substituted.
  • substituted means that a hydrogen atom is removed and replaced by a substituent. It is to be understood that substitution at a given atom is limited by valency.
  • Cn-m alkyl refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbon atoms. In some embodiments, the alkyl group contains 1 to 6, or 1 to 3 carbon atoms.
  • alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, /e/V-butyl , «-pentyl, 2-methyl-l-butyl, 3-pentyl, «-hexyl, 1,2,2- trimethylpropyl, and the like.
  • alkylene employed alone or in combination with other terms, refers to a divalent alkyl linking group, which can be branched or straight-chain, where the two substituents may be attached any position of the alkylene linking group.
  • alkylene groups include, but are not limited to, ethan-l,2-diyl, propan- 1, 3 -diyl, propan- 1,2-diyl, and the like.
  • HO-Ci-3-alkyl refers to a group of formula - alkylene-OH, wherein said alkylene group has 1 to 3 carbon atoms.
  • CN-C1-3 alkyl refers to a C1-3 alkyl substituted by a cyano group.
  • amino refers to a group of formula -NH2.
  • di(Ci-3-alkyl)amino refers to a group of formula - N(alkyl)2, wherein the two alkyl groups each has, independently, 1 to 3 carbon atoms.
  • C1-3 alkylamino refers to a group of formula -NH(alkyl), wherein the alkyl group has 1 to 3 carbon atoms.
  • di(Ci- 3 alkyl)aminosulfonyl refers to a group of formula -S(0)2N(alkyl)2, wherein each alkyl group independently has 1 to 3 carbon atoms.
  • C1-3 alkyl sulfonyl refers to a group of formula -S(0)2-alkyl, wherein the alkyl group has 1 to 3 carbon atoms.
  • halo or “halogen”, employed alone or in combination with other terms, includes fluoro, chloro, bromo, and iodo. In some embodiments, the halo group is fluoro or chloro.
  • Cn-mhaloalkyl refers to a Cn-m alkyl group having up to ⁇ 2(n to m)+l ⁇ halogen atoms which may either be the same or different.
  • the halogen atoms are fluoro atoms.
  • the alkyl group has 1-6 or 1-3 carbon atoms.
  • Example haloalkyl groups include CF3, C2F5, CHF2, CCh, CHCI2, C2CI5, and the like.
  • the haloalkyl group is a fluoroalkyl group.
  • C1-3 fluoroalkyl refers to a C1-3 alkyl group that may be partially or completely substituted by fluoro atoms.
  • C3-6 cycloalkyl refers to a non-aromatic monocyclic hydrocarbon moiety, having 3- 6 carbon atoms, which may optionally contain one or more alkenylene groups as part of the ring structure.
  • One or more ring-forming carbon atoms of a cycloalkyl group can be oxidized to form carbonyl linkages.
  • Exemplary C3- 6 cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, and the like.
  • the cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • C3- 6 cycloalkyl-Ci- 3 alkyl refers to a group of formula -C1- 3 alkylene- C 3 - 6 cycloalkyl.
  • the compounds described herein can be asymmetric ( e.g ., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Compounds that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
  • An example method includes fractional recrystallization using a chiral resolving acid which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as b-camphorsulfonic acid.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a-methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N- methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g ., dinitrobenzoylphenylglycine).
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. For example, it will be recognized that the following pyrazole ring may form two tautomers:
  • All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g. hydrates and solvates) or can be isolated.
  • the compounds described herein, or salts thereof are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the compounds described herein.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds described herein, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • ambient temperature and “room temperature” or “rt” as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20 °C to about 30 °C.
  • the present application also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present application include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present application can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • ACN acetonitrile
  • the term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • “contacting” a JAK with a compound of the invention includes the administration of a compound of the present application to an individual or patient, such as a human, having a JAK, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the JAK.
  • the inhibitors are administered in a therapeutically effective amount.
  • therapeutically effective amount refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician. .
  • treating refers to one or more of (1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); (2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease; or (3) preventing the disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease.
  • treating refers to inhibiting or ameliorating the disease.
  • treating is preventing the disease.
  • the methods described herein can further comprise administering one or more additional therapeutic agents.
  • the one or more additional therapeutic agents can be administered to a patient simultaneously or sequentially.
  • the additional therapeutic agent is an antibiotic.
  • the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, erythromycin, metronidazole, rifampin, moxifloxacin, dapsone, or a combination thereof.
  • the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, or erythromycin in combination with metronidazole.
  • the antibiotic is a combination of rifampin, moxifloxacin, and metronidazole.
  • the antibiotic is a combination of moxifloxacin and rifampin.
  • the additional therapeutic agent is a retinoid.
  • the retinoid is etretinate, acitretin, or isotretinoin.
  • the additional therapeutic agent is a steroid. In some embodiments, the additional therapeutic agent is a corticosteroid. In some embodiments, the steroid is such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisone, prednisolone, or flumetholone.
  • the additional therapeutic agent is an anti-TNF-alpha agent.
  • the anti-TNF-alpha agent is an anti-TNF-alpha antibody.
  • the anti-TNF-alpha agent is infliximab or etanercept, or adalimumab.
  • the additional therapeutic agent is an immunosuppressant.
  • the immunosuppressant is methotrexate or cyclosporin A.
  • the immunosuppressant is mycophenolate mofetil or mycophenolate sodium.
  • the additional therapeutic agent is finasteride, metformin, adapalene or azelaic acid.
  • the method further comprises administering an additional therapeutic agent selected from IMiDs, an anti-IL-6 agent, a hypomethylating agent, and a biologic response modifier (BRM).
  • an additional therapeutic agent selected from IMiDs, an anti-IL-6 agent, a hypomethylating agent, and a biologic response modifier (BRM).
  • a BRM is a substances made from living organisms to treat disease, which may occur naturally in the body or may be made in the laboratory.
  • BRMs include IL-2, interferon, various types of colony-stimulating factors (CSF, GM-CSF, G-CSF), monoclonal antibodies such as abciximab, etanercept, infliximab, rituximab, trasturzumab, and high dose ascorbate.
  • the hypomethylating agent is a DNA methyltransferase inhibitor.
  • the DNA methyltransferase inhibitor is selected from 5 azacytidine and decitabine.
  • IMiDs are as immunomodulatory agents.
  • the IMiD is selected from thalidomide, lenalidomide, pomalidomide, CC-11006, and CC-10015.
  • the method further comprises administering an additional therapeutic agent selected from anti -thymocyte globulin, recombinant human granulocyte colony-stimulating factor (G CSF), granulocyte-monocyte CSF (GM-CSF), an erythropoiesis-stimulating agent (ESA), and cyclosporine.
  • an additional therapeutic agent selected from anti -thymocyte globulin, recombinant human granulocyte colony-stimulating factor (G CSF), granulocyte-monocyte CSF (GM-CSF), an erythropoiesis-stimulating agent (ESA), and cyclosporine.
  • the method further comprises administering an additional JAK inhibitor to the patient.
  • the additional JAK inhibitor is selected from a JAK2 inhibitor (e.g, a selective JAK2 inhibitor), a JAK1/JAK2 inhibitor, a JAK3 inhibitor (e.g, a selective JAK3 inhibitor), and a JAK1/JAK3 inhibitor, or a pharmaceutically acceptable salt of any of the aforementioned.
  • the additional JAK inhibitor is a JAK1/JAK2 inhibitor.
  • the JAK1/JAK2 inhibitor is selective for JAK1 and JAK2 over JAK3 and TYK2.
  • the JAK1/JAK2 inhibitor is 3-cyclopentyl-3- [4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]propanenitrile, or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is (3R)-3-cyclopentyl-3-[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljpropanenitrile (ruxolitinib), or a pharmaceutically acceptable salt thereof.
  • Ruxolitinib has an ICso of less than 10 nM at 1 mM ATP at JAK1 and JAK2.
  • the phosphoric acid salt can be made as described in U.S. Patent 8,722,693, which is incorporated herein by reference in its entirety.
  • the JAK1/JAK2 inhibitor is barcitinib, or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is barcitinib.
  • the additional JAK1 inhibitor is a JAK1/JAK3 inhibitor.
  • the JAK1/JAK3 inhibitor is tofacitinib, or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK3 inhibitor is tofacitinib.
  • the JAK1/JAK2 inhibitor can be an isotopically-labeled compound, or a pharmaceutically acceptable salt thereof.
  • An “isotopically” or “radio- labeled” compound is a compound wherein one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present disclosure include but are not limited to 2 H (also written as D for deuterium), 3 H (also written as T for tritium), U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 18 F, 35 S, 36 C1, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 124 I, 125 I and 131 I.
  • one or more hydrogen atoms in a compound of the present disclosure can be replaced by deuterium atoms, such as -CD3 being substituted for -CFb).
  • the JAK1/JAK2 inhibitor is a compound, wherein one or more hydrogen atoms in the compound are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is ruxolitinib, wherein one or more hydrogen atoms are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is any of the compounds in US Patent 9,249,149, which is incorporated herein by reference in its entirety, or a pharmaceutically acceptable salt thereof.
  • the inhibitor of JAK1/JAK2 is CTP-543, or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is a compound of Formula IV: or a pharmaceutically acceptable salt thereof, wherein:
  • R 4 is selected from H and D; each R 5 is the same and is selected from H and D; and
  • R 6 , R 7 , and R 8 are each independently selected from H and D; provided that when R 1 is H, each R 2 and each R 3 are H, R 4 is H, and each of R 6 , R 7 , and R 8 is H, then each R 5 is D.
  • the JAK1/JAK2 inhibitor is a compound of Formula IV selected from the following compounds 100-130 in the table below (wherein R 6 , R 7 , and R 8 are each H), or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is a compound of Formula IV selected from the following compounds 200-231 in the table below (wherein R 6 , R 7 , and R 8 are each D), or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is baricitinib, wherein one or more hydrogen atoms are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof.
  • the JAK1/JAK2 inhibitor is any of the compounds in US Patent 9,540,367 (which is incorporated herein by reference in its entirety), or a pharmaceutically acceptable salt thereof.
  • the additional JAK inhibitor is selected from baricitinib, tofacitinib, oclacitinib, filgotinib, gandotinib, lestaurtinib, momelotinib, bacritinib, PF-04965842, upadacitinib, peficitinib, fedratinib, cucurbitacin I, ATI-501 (Aclaris), ATI-502 (Aclaris), JTE-052 (z.e., delgocitinib; Leo Pharma and Japan Tobacco), and CHZ868.
  • Additional agents including, but not limited to, anti-inflammatory agents, immunosuppressants, PI3K6 inhibitors, mTor inhibitors, Bcr-Abl inhibitors, Flt-3 inhibitors, RAF inhibitors, and FAK kinase inhibitors (e.g ., those described in WO 2006/056399, which is incorporated herein by reference in its entirety), or other agents can be used in combination with the JAK1 inhibitors described herein for treatment of cutaneous lupus erythematosus (CLE).
  • the one or more additional agents can be administered to a patient simultaneously or sequentially.
  • Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No.
  • Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120, all of which are incorporated herein by reference in their entirety.
  • Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444, both of which are incorporated herein by reference in their entirety.
  • Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402, all of which are incorporated herein by reference in their entirety.
  • one or more of the JAK1 inhibitors described herein can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • the additional therapeutic agent is fluocinolone acetonide (Retisert®) or rimexolone (AL-2178, Vexol, Alcon).
  • the additional therapeutic agent is cyclosporine (Restasis®).
  • the additional therapeutic agent is selected from DehydrexTM (Holies Labs), Civamide (Opko), sodium hyaluronate (Vismed, Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine, doxycycline (ALTY-0501, Alacrity), minocycline, iDestrinTM (NP50301, Nascent Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline (Duramycin, MOLI1901, Lantibio), CF101 (2S,3S,4R
  • the additional therapeutic agent is an anti -angiogenic agent, cholinergic agonist, TRP-1 receptor modulator, a calcium channel blocker, a mucin secretagogue, MUC1 stimulant, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, or an mTOR inhibitor.
  • the additional therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycline).
  • the additional therapeutic agent binds to FKBP12.
  • the additional therapeutic agent is an alkylating agent or DNA cross-linking agent; an anti-metabolite/demethylating agent (e.g., 5- flurouracil, capecitabine or azacitidine); an anti -hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e.g. vincristine or paclitaxel); an topoisomerase (I or II) inhibitor (e.g. mitoxantrone and irinotecan); an apoptotic inducers (e.g. ABT-737); a nucleic acid therapy (e.g.
  • an anti-metabolite/demethylating agent e.g., 5- flurouracil, capecitabine or azacitidine
  • an anti -hormone therapy e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor
  • a mitotic inhibitor e.g. vincristine or paclit
  • RNAi nuclear receptor ligands
  • nuclear receptor ligands e.g., agonists and/or antagonists: all- trans retinoic acid or bexarotene
  • epigenetic targeting agents such as histone deacetylase inhibitors (e.g. vorinostat), hypomethylating agents (e.g. decitabine); regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
  • the additional therapeutic agent is selected from an antibiotic, an antiviral, an antifungal, an anesthetic, an anti-inflammatory agent (e.g., steroidal and non-steroidal anti-inflammatory agents), and an anti-allergic agent.
  • suitable agents include, but are not limited to, aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine; sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin; colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives (“rifampins”); cycloserine; beta
  • the JAK1 inhibitors provided herein can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including intradermal, transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g, by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • topical including intradermal, transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery
  • pulmonary e.g, by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g, intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, dermal patches, solutions, suspensions, foams, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids, and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • the composition is formulated for topical administration by transdermal patch, dermal patch, solution, suspension, gel, cream, ointment, lotion, spray, foam, liquid, drops, suppository, and powder.
  • the composition is formulated for topical administration by transdermal patch.
  • the composition is formulated for topical administration by dermal patch.
  • the composition is formulated as a foam ( e.g ., for topical administration).
  • topical drugs which are able to penetrate the skin barrier and provide limited systemic effects are of particular importance.
  • Topical (dermal/intradermal) formulations are typically solutions, suspensions, gels, creams, ointments, lotions, sprays and foams.
  • Preferred topical formulations should be physically and chemically stable, not cause skin irritation, and deliver the active agent (e.g., a selective JAK1 inhibitor, or a pharmaceutically acceptable salt thereof, as described herein) at the appropriate layer of the skin in concentrations that would result in therapeutic response, with limited systemic exposure.
  • the administration is topical and comprised of formulations with one or more pharmaceutically (e.g, dermatologically) acceptable excipients.
  • dermatologically acceptable excipients include, but are not limited to, a pH adjusting agents, chelating agents, preservatives, co-solvents, penetration enhancers, humectants, thickening, gelling, viscosity building agents, surfactants, propellants, fragrance, colorants, or any combination or mixture thereof.
  • the topical formulation is administered locally to the patient (e.g, administered at the site of a lesion).
  • the pH-adjusting agent is selected from an acid, an acid salt, a base, a base salt, and a buffer, or any mixture thereof.
  • Exemplary acids include, but are not limited to, lactic acid, acetic acid, citric acid, and benzoic acid, and salts thereof.
  • Exemplary bases include, but are not limited to, trolamine, tromethamine, and salts thereof.
  • Exemplary buffers include, but are not limited to, citrate/citric acid, acetate/acetic acid, edetate/edetic acid, lactate/lactic acid, and the like.
  • the chelating agent is a single excipient. In some embodiments, the chelating agent is a mixture of two or more chelating agents. Exemplary chelating agents include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), or a salt thereof. In some embodiments, the chelating agent comprises a mixture of a chelating agent and an antioxidant, wherein the chelating agent and antioxidant prevent, minimize, or reduce oxidative degradation reactions in the composition. Exemplary anti-oxidants include, but are not limited to, butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), tocopherol, and propyl gallate.
  • BHT butylated hydroxytoluene
  • BHA butylated hydroxyanisole
  • tocopherol and propyl gallate.
  • the composition comprises one or more preservatives. In some embodiments, the composition comprises a mixture of two or more preservatives. In some embodiments, the composition comprises one to five preservatives. Exemplary preservatives include, but are not limited to, benzyl alcohol, phenonyexthanol, methyl paraben, ethyl paraben, propyl paraben, butyl paraben, and imidazolidinyl urea.
  • the composition comprises one or more co-solvents. In some embodiments, the composition comprises a mixture of two or more co-solvents. In some embodiments, the composition comprises one to five co-solvents.
  • Exemplary solvents include, but are not limited to, water, propylene glycol, diethylene glycol monoethyl ether, dimethyl isosorbide, ethyl alcohol, isopropyl alcohol, benzyl alcohol, propanediol, propylene glycol, polyethylene glycols ( e.g ., polyethylene glycol 200, polyethylene glycol 300, polyethylene glycol 400, and the like).
  • the solvent is a non-water soluble agent. Exemplary non-water soluble agents include, but are not limited to, diethyl sebacate, diisopropyl adipate, isopropyl myristate, isopropyl palmitate, and medium chain triglycerides.
  • the composition comprises one or more penetration enhancers. In some embodiments, the composition comprises a mixture of two or more penetration enhancers. In some embodiments, the composition comprises one to five penetration enhancers.
  • the penetration enhancers can act as both a solvent and a penetration enhancer. Exemplary penetration enhancers include, but are not limited to, fatty acids, fatty acid esters, fatty alcohols, pyrrolidones, sulfoxides, alcohols, diols and polyols, or any mixture thereof.
  • a co-solvent provided herein is a penetration enhancer.
  • the composition comprises one or more thickening, gelling, or viscosity building agents. In some embodiments, the composition comprises a mixture of two or more thickening, gelling, or viscosity building agents. In some embodiments, the composition comprises one to five thickening, gelling, or viscosity building agents.
  • Exemplary thickening, gelling, or viscosity building agents include, but are not limited to, cellulosic derivatives (e.g ., hydroxy ethylcellulose (HEC), carboxymethylcellulose, hydroxypropylcellulose (HPC), and hydroxypropyl methylcellulose (HPMC), and polyvinylpyrrolidone (PVP).
  • the surfactant is a compound that lowers the surface tension between two liquids or between a liquid and a solid.
  • Surfactant may be a mixture of two or more surfactants.
  • Exemplary surfactants include, but are not limited to, ethoxylated fatty alcohol ether (e.g., steareth-2, steareth-10, steareth-20, ceteareth-2, ceteareth-10, and the like), PEG esters (e.g., PEG-4 dilaurate, PEG-20 stearate, and the like), Glyceryl esters or derivatives thereof (e.g, glyceryl dioleate, glyceryl stearate, and the like), polymeric ethers (e.g, poloxamer 124, poloxamer 181, poloxamer 182, and the like), sorbitan derivatives (e.g, polysorbate 80, sorbitan monostearate, and the like), fatty alcohols (e.g, cet
  • the administration is topical administration to the skin.
  • the administration is oral.
  • administration of a topical composition comprising a selective JAK1 inhibitor provided herein to a patient provides a systemic exposure (Cmax) at steady state of about 5% or less of the applied dose, e.g, about 4% or less, about 3% or less, about 2% or less, about 1% or less, and the like.
  • administration of a topical composition comprising a selective JAK1 inhibitor provided herein to a patient provides a systemic exposure (Cmax) at steady state of about 1% or less of the applied dose
  • This invention also includes pharmaceutical compositions which contain, as the active ingredient, a JAK1 inhibitors provided herein, or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • JAK1 inhibitors provided herein may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types.
  • Finely divided (nanoparticulate) preparations of the JAK1 inhibitors can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • the pharmaceutical composition comprises silicified microcrystalline cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof.
  • SMCC silicified microcrystalline cellulose
  • the silicified microcrystalline cellulose comprises about 98% microcrystalline cellulose and about 2% silicon dioxide w/w.
  • the composition is a sustained release composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
  • the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one component selected from microcrystalline cellulose, lactose monohydrate, hydroxypropyl methylcellulose, and polyethylene oxide.
  • the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate, and hydroxypropyl methylcellulose.
  • the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate, and polyethylene oxide.
  • the composition further comprises magnesium stearate or silicon dioxide.
  • the microcrystalline cellulose is Avicel PHI 02TM.
  • the lactose monohydrate is Fast-flo 316TM.
  • the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M PremierTM) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel K00LVTM).
  • the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Poly ox WSR 1105TM).
  • a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 1 to about 1,000 mg, from about 1 mg to about 100 mg, from 1 mg to about 50 mg, and from about 1 mg to 10 mg of active ingredient.
  • the dosage is from about 1 mg to about 50 mg or about 1 mg to about 10 mg of active ingredient.
  • each dosage contains about 10 mg of the active ingredient.
  • each dosage contains about 50 mg of the active ingredient.
  • each dosage contains about 25 mg of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the compositions comprise from about 1 to about 1,000 mg, from about 1 mg to about 100 mg, from 1 mg to about 50 mg, and from about 1 mg to 10 mg of active ingredient.
  • the compositions comprise from about 1 mg to about 50 mg or about 1 mg to about 10 mg of active ingredient.
  • this embodies compounds or compositions containing about 1 mg to about 10 mg, about 1 mg to about 20 mg, about 1 mg to about 25 mg, about 1 mg to about 50 mg of the active ingredient.
  • the active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present application.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present application.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present application (e.g. a JAK1 inhibitor provided herein).
  • the tablets or pills of the present application can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the compounds and compositions of the present application can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical (e.g, intradermal) administration provides the advantage of treating the skin disorder locally, minimizing potential adverse events associated with systemic exposure, and allowing an easier discontinuation of the therapy, if necessary. Additionally, some topical dosage forms such as creams, ointments, and gels have the benefit of excipients that may act as emollients or occlusive agents, which can increase patient well-being and compliance during the treatment period. Other dosage routes such as oral, parenteral, and inhalation may lead to supratherapeutic systemic drug levels, increased likelihood of adverse events, drug-drug interactions, and generation of active/toxic metabolites, which may result in treatment discontinuation and inadequate patient compliance.
  • Topical formulations intended for dermal delivery are typically solutions, suspensions, gels, creams, ointments, lotions, sprays, and foams and can contain one or more conventional carriers as described herein.
  • the formulation composition should be prepared with the goal of delivering the active ingredient to the appropriate layer(s) of the skin, minimizing systemic exposure, and preventing skin irritation. Additionally the pharmaceutical composition should be physically and chemically stable.
  • one or more additional excipients as described herein may be necessary, e.g ., pH adjusting agents, chelating agents, preservatives, co-solvents, penetration enhancers, humectants, thickening, gelling, viscosity building agents, surfactants, propellants, fragrances, colorants, or any combination or mixture thereof.
  • topical formulations can contain one or more conventional carriers as described herein.
  • ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white petrolatum, and the like.
  • Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g. glycerinemonostearate, PEG- glycerinemonostearate and cetylstearyl alcohol.
  • Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention.
  • the topical formulations can be suitably packaged in tubes of, for example, 100 g, which are optionally associated with instructions for the treatment of cutaneous lupus erythematosus (CLE).
  • CLE cutaneous lupus erythematosus
  • compositions can be administered to a patient already suffering from cutaneous lupus erythematosus (CLE) in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • CLE cutaneous lupus erythematosus
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the therapeutic dosage of a compound of the present application can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g ., hydrophobicity), and the route of administration.
  • the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 pg/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the compositions of the invention can further include one or more additional pharmaceutical agents, examples of which are listed hereinabove.
  • kits useful for example, in the treatment and/or prevention of cutaneous lupus erythematosus (CLE), which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a JAK1 inhibitor described herein.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • JAK1 inhibitors that can be used for the treatment of cytokine-related diseases or disorders are tested for inhibitory activity of JAK targets according to the following in vitro assay described in Park el al. , Analytical Biochemistry 1999, 269 , 94-104.
  • JAK1 The catalytic domains of human JAK1 (a.a. 837-1142), JAK2 (a. a. 828-1132) and JAK3 (a.a. 781-1124) with an N-terminal His tag are expressed using baculovirus in insect cells and purified.
  • the catalytic activity of JAK1, JAK2 or JAK3 was assayed by measuring the phosphorylation of a biotinylated peptide.
  • the phosphorylated peptide was detected by homogenous time resolved fluorescence (HTRF).
  • ICsos of compounds are measured for each kinase in the 40 microL reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8) buffer with 100 mM NaCl, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA.
  • ATP concentration in the reactions is 1 mM.
  • Reactions are carried out at room temperature for 1 hour and then stopped with 20 pL 45 mM EDTA, 300 nM SA-APC, 6 nM Eu- Py20 in assay buffer (Perkin Elmer, Boston, MA).
  • NGS Next Generation Sequencing
  • Illumina HiSeq 2500 was used for RNA sequencing (Standard 3’RNA seq with 50 cycles)
  • RNA sequencing Standard 3’RNA seq with 50 cycles
  • pJAKl was significantly increased in keratinocytes from stratum basale to stratum granulosum and in dermal infiltrating immune cells. It was also observed that pJAKl was significantly enhanced in Lichen planus, an autoimmune disease sharing common histological features with CLE.
  • Immortalized keratinocytes (HaCaT), were acquired from CLS Cell Lines Service GmbH, Eppelheim, Germany), normal human epidermal keratinocytes (NHEKs, FC-0025) and Human epidermis equivalents (epiCS, CS-1001) from CellSystems, Troisdorf, Germany. These cell lines were cultured according the manufactures protocols. Cultured keratinocytes were stimulated with endogenous nucleic acids (eNA, 1,25 pg/mL) isolated from unstimulated keratinocytes using the “Genomic DNA from tissue“ kit (Machery- Nagel, Dueren, Germany).
  • eNA endogenous nucleic acids
  • Lipofectamine 2000 (Invitrogen, Carlsbad, USA) functioned as a transfection reagent (2.5 pL/mL).
  • Enzyme-linked immunosorbant assays for human CXCL10 were performed using DuoSet Ancillary Reagent Kit 2 (DY008 R&D systems) according to the supplied protocol, measured by Synergy HT Multi- Detection Multiplate Reader (BioTek, Winooski, VT, USA) and read out with Gen5 software (version 1.11.5).
  • JAKl-phosphorylation was strongly enhanced within immortalized keratinocytes (HaCaT) after stimulation with eNA, corresponding with the findings in CLE skin lesions described above (see Examples 2-3).
  • HaCaT immortalized keratinocytes
  • JAKl-specific INCB039110 and JAK 1/2-specific ruxolitinib significantly decreased the activation of JAKl within stimulated cells.
  • KEGG pathways were classified using Database for Annotation, Visualization and Integrated Discovery (DAVID ver. 6.8). E-values were generated with EASE Score. Count: number of genes >2- fold downregulated in NHEK by INCB039110 within the respective KEGG pathway.
  • JAKl selective inhibitors are as potent as JAK1/2 inhibitors in suppression of CLE typical cytokines, as shown in FIG. 2D, and prohibit gene expression encoding proinflammatory chemokines (CXCLlO-11), lymphocyte activators (BLyS) (see e.g. , Wenzel et al, Exp. Dermatol. 2018, 27:95-97) and cell death promotors (TRAIL (see e.g., Zahn et al, Br. J. Dermatol. 2011, 165:1118-1123), AIM2, Caspase 10, TREX1).
  • inhibition of JAK3 did not obtain a reduction of CXCL10 expression, as shown in FIG.
  • TREX1 mice (generated on C57BL/6J background; Cancer Research Institute, London, UK) were bred and maintained under specific pathogen-free conditions at the animal core facility of UKB Bonn (HET, Bonn, Germany).
  • TREX1 _/ mice spontaneously developed CLE-like erythrosquamous and partly ulcerated skin lesions at a certain age which intensified after UVB -provocation.
  • distinct histological features such as epidermal thickness and infiltrating dermal immune cells were significantly improved by JAK1 inhibition, as shown in FIG. 3C.

Abstract

The present application provides methods of treating a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a JAK1 selective inhibitor, or a pharmaceutically acceptable salt thereof.

Description

USE OF JAK1 INHIBITORS FOR THE TREATMENT OF CUTANEOUS LUPUS ERYTHEMATOSUS AND LICHEN PLANUS (LP)
TECHNICAL FIELD
The present application provides methods for the treatment of cutaneous lupus erythematosus and/or Lichen planus (LP) using compounds that modulate the activity of Janus kinase (JAK) 1.
BACKGROUND
Cutaneous lupus erythematosus (CLE) is an inflammatory autoimmune skin disease with heterogenic subtypes varying from localized discoid plaques to severe and widespread erythrosquamous lesions in affected patients (see e.g, Kuhn & Landmann, J. Autoimmun. 2014, 48-49:14-19). The disease is characterized by a typical histological pattern called “interface dermatitis” (ID) consisting of colloid bodies and an anti-epidermal cytotoxic lymphocytic infiltrate in the dermoepidermal junction which is orchestrated by IFN-regulated proinflammatory cytokines (see e.g. , Wenzel & Tilting, J. Invest. Dermatol. 2008, 128:2392-2402; and Wenzel et al, Br. J. Dermatol. 2007, 157:752-757).
CLE is of multifactorial origin; including both genetic and environmental risk factors (see e.g., Kunz et al, Exp. Dermatol. 2015, 24:510-515; Sinha & Dey-Rao, J. Investig. Dermatol. Symp. Proc., 2017, 18:S75-S80; and Szczqch et al, Lupus , 2017, 26:791-807). In particular UV radiation promotes cellular damage, apoptosis and release of DNA-containing blebs. It is considered that cell debris clearance is impaired entailing secondary necrosis (see e.g., Kuhn et al, Arthritis Rheum. 2006, 54:939-950; Caricchio et al, The Journal of Immunology 2003 , 171:5778-5786; and Mahajan et al, Front Immunol. 2016, 7). In some cases such as familial chilblain lupus the underlying cause can be TREX1 gene mutations leading to a dysfunctional TREX1 exonuclease and high accumulation of cytosolic DNA (see e.g. , Giinther et al, J. Am. Acad. Dermatol. 2013, 69:el79-81; Grieves et al, Proc. Natl. Acad. Sci. U.S.A. 2015, 112:5117-5122; Giinther et al, Dermatology (Basel), 2009, 219:162-166; and Peschke et al, J. Invest. Dermatol. 2014, 134:1456-1459). When nuclear components, such as endogenous nucleic acid motifs, are released out of the nucleus due to cellular damage, they can be perceived as danger associated molecular patterns (DAMPs) (see e.g, Scholtissek et al, J. Invest. Dermatol. 2017, 137:1484-1492). There is evidence that keratinocytes and particularly plasmacytoid dendritic cells (pDCs) react on DAMPs inappropriately with immense type I IFN production through TLR-dependent or TLR-independent pathways in CLE (see e.g., Mustelin et al, Front. Immunol. 2019, 10:238; Wollenberg et al, J. Invest. Dermatol. 2002, 119: 1096-1102; Wenzel et al, Arch. Dermatol. Res. 2009, 301:83-86; Yu et al, J. Autoimmun. 2013, 41:34-45; and Katayama et al, J. Invest. Dermatol. DOI: 10.1016/j jid.2019.02.035). Undergoing an autocrine loop IFNs bind to IFN-a/b receptors on keratinocytes, thus activating JAK/STAT pathway and expression of proinflammatory mediators such as CXCL10. It is known that CXCL10 recruits CXCR3+ effector cells and pDCs into skin lesions (see e.g. , Wenzel et al, Arch. Dermatol. Res. 2009, 301:83-86; and Wenzel et al, J. Pathol. 2005, 205:435-442). Those effector cells induce keratinocytic necroptosis (see e.g , Lauffer et al, J. Invest. Dermatol. 2018, 138:1785-1794), cytokine release and a continuous “self-recruitment” as a hallmark of chronic inflammation (see e.g. , Meller et al, Arthritis Rheum. 2005, 52:1504-1516).
This circle can result in a strong burden for quality of life (see e.g. , Samotij et al, Postepy Dermatol. Alergol. 2018, 35:192-198; and Ogunsanya et al, Int. J.
Womens Dermatol. 2018, 4:152-158). Following current guidelines, corticosteroids and antimalarials are established as first-line treatment of CLE. However, corticosteroids are limited by side effects and long-term treatment, particularly in antimalarial-resistant patients, can be challenging. In addition, no specifically approved drugs exist for CLE and only a few clinical trials have been conducted, not least because of clinical heterogeneity and thus challenging trial design (see e.g. ,
Chen et al, FlOOORes. 2019, 8).
The American College of Rheumatology (ACR) has established a scheme of eleven clinical and lab criteria for the purpose of distinguishing Systemic Lupus Erythematosus (SLE) from other autoimmune diseases. The ACR guidelines require four of eleven criteria to be met for a diagnosis of SLE, however, only four of the criteria are cutaneous in nature (malar rash, discoid lesions, mucosal ulcers, and photosensitivity) and therefore involvement of skin pathologies is not obligatory for the diagnosis of SLE. Conversely, cutaneous lupus erythematosus (CLE) subtypes can occur in the absence of systemic disease (SLE); CLE is two to three times more frequent than SLE (see e.g ., Tebbe & Orfanos, Lupus , 1997, 6(2):96-104).
Considering these limitations, there is a medical need for new therapeutic options. This application is directed to that need and others.
SUMMARY
The present application provides methods of treating a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a JAK1 selective inhibitor provided herein.
DESCRIPTION OF DRAWINGS
FIGs. 1 A-1D show the functional role of JAK1 expression in CLE skin lesions. FIG. 1 A shows representative immunohistochemical sections (anti- phosphoJAKl staining) from skin samples of patients with different inflammatory skin disorders (subacute cutaneous lupus erythematosus (SCLE, n=5), chronic discoid lupus erythematosus (CDLE, n=5), Lichen planus (LP, n=6) and healthy controls (HC, n=5)), original magnification x200. FIG. IB shows the expression of pJAKl, scored semiquantitatively by scoring from 0 = no expression to 3 = strong expression (see e.g ., Wenzel et al. J Pathol. 2005, 205:435-442). All bars show mean ± SEM, td = p <0,1, * = p <0,05, ** = p <0,01 (Kruskal -Wallis-Test). FIG. 1C shows upregulated expression of IFN-associated genes in CLE lesional skin (2-fold, / <0,01 ; Welch’s /-test) compared to healthy controls (HC). FIG. ID shows KEGG pathways upregulated in CLE. KEGG pathway classification was performed using Database for Annotation, Visualization and Integrated Discovery (DAVID ver. 6.8). C-values were generated with EASE Score. Count: number of genes >2-fold upregulated in CLE within the respective KEGG pathway.
FIGs. 2A-2E show the effects of pharmacological JAK1 inhibition on proinflammatory cytokine expression and IFN-associated gene regulation in cultured keratinocytes. FIG. 2A shows mean area intensity in pixel values of anti-pJAKl- antibody staining in stimulated HaCaT (eNA), stimulated and inhibitor-treated HaCaT (eNA+ JAKl inhibitor {l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4- yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3- yl } acetonitrile (i.e., INCB039110); eNA+ JAKl/2, inhibitor: ruxolitinib) and unstimulated HaCaT (C-Medium). Measurement was performed out using Fiji software. Measurements per entity: n=5. All scatter dot plots show mean + SEM, **
= p<0,01 (Mann-Whitney U test). FIG. 2B shows representative immunofluorescence micrographs of eNA-stimulated untreated (eNA) or inhibitor-treated (eNA+JAKl, inhibitor:INCB039110; eNA+JAKl/2, inhibitonruxolitinib) HaCaT as well as unstimulated HaCaT-cells (Medium Control). Anti-pJAKl antibody staining in red, DAPI staining of the nucleus in blue, original magnification x400. FIG. 2C shows expression of CLE-typical genes within eNA-stimulated NHEK (+, 4 samples) and >2-fold downregulated expression (p< 0,05; Welch's /-test) after treatment of stimulated NHEK with JAKl selective INCB039110 (JAKl; 4 samples). FIG. 2D shows the effect of different JAK inhibitors on CXCL10 expression in stimulated HaCaT after 24 hours of incubation. Cells were treated with JAKl selective INCB039110 (JAKl), JAKl/2 selective ruxolitinib (JAK 1/2) and JAK3 selective FM- 381 (JAK3). Measured by ELISA. All bars show mean + SEM, *** = K0,001, ** = p< 0,01, td = / 0, 1 (Mann-Whitney U test). FIG. 2E shows micrographs representing the expression of CXCL10 in stimulated cells (eNA, n=3) and JAKl inhibitor treated cells (INCB039110) after stimulation (eNA+JAKl, n=3), original magnification x400, with corresponding CXCL10 expression score. All bars show mean ± SEM, *
= p<0,05; (Mann-Whitney U test).
FIGs. 3A-3C show pharmacological JAKl inhibition in vivo in a lupus-prone TREX1 mouse model. FIG. 3 A shows clinical findings of UV-stimulated (started on day -3; 3x450 mJ/cm2 for 115 seconds) TREXl mice (n=8) on baseline (day 0, start of treatment) and after 4 and 7 days of treatment with JAKl selective INCB039110 or placebo. FIG. 3B shows mean score of the mouse-adapted revised CLE-disease-area- and-severity index (Mouse RCLASI) ± SEM in n=4 mice per group. FIG. 3C shows histological (H&E) and immunohistological (CD45) micrographs of lesional skin, original magnification x400. Corresponding inflammation score of CD45 expression in lesional skin of placebo-treated and JAK1 -inhibitor treated mice. All bars show mean ± SEM, ** = p< 0,01 (Mann-Whitney U test).
DETAILED DESCRIPTION
The present application provides, inter alia , methods of treating a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering a therapeutically effective amount of JAK1 selective inhibitor, or a pharmaceutically acceptable salt thereof. In some embodiments, the disease is cutaneous lupus erythematosus (CLE). In some embodiments, the cutaneous lupus erythematosus (CLE) is selected from acute cutaneous lupus erythematosus, subacute cutaneous lupus erythematosus, and chronic cutaneous lupus erythematosus. In some embodiments, the cutaneous lupus erythematosus (CLE) is acute cutaneous lupus erythematosus. In some embodiments, the cutaneous lupus erythematosus is selected from subacute cutaneous lupus erythematosus (SCLE) and chronic discoid lupus erythematosus (CDLE). In some embodiments, the cutaneous lupus erythematosus is subacute cutaneous lupus erythematosus (SCLE). In some embodiments, the cutaneous lupus erythematosus is chronic discoid lupus erythematosus (CDLE). In some embodiments, the disease is Lichen planus (LP).
As used herein, “cutaneous lupus” refers to a form of the disease in which symptoms are restricted to those that affect the skin. For example, a patient may be diagnosed with cutaneous lupus, but that does not mean that he or she has SLE, which affects multiple parts of the body. Similarly, if a patient has SLE, it does not mean that he or she will necessarily have cutaneous lupus. Accordingly the Cutaneous Lupus Area and Severity Index (CLASI) scoring system was devised, which quantitatively measures disease activity and damage (see e.g ., Albrecht et ah, . ./. Invest. Dermatol. 2005, Nov; 125(5):889-94). This index, which accounts for lesional morphology as well as anatomic location, has since been validated by reliability testing for both dermatologists and rheumatologists.
Cutaneous lupus is divided into several subtypes, including acute cutaneous lupus erythematosus, subacute cutaneous lupus erythematosus, and chronic cutaneous lupus erythematosus. Acute cutaneous LE typically presents in the third decade of life and is frequently associated with active SLE. The most typical form of acute cutaneous lupus is a malar rash-flattened areas of red skin on the face that resemble a sunburn (see e.g. , Fabbri et al., Am. ./. Clin. Dermatol. 2003, 4(7):449-65). These lesions are typically transient, sun-induced, and non-scarring. Malar rashes have been reported to be present in -50% of SLE patients at the time of diagnosis, with clinical activity of the rash paralleling that of the systemic disease (see e.g. , Rothfield et al., Clin. Dermatol .; 2006, Sep-Oct; 24(5):348-62).
Chronic cutaneous lupus includes discoid LE (DLE), LE profundus (LEP), chilblain LE (CELLE), and LE tumidus (LET). In some embodiments, the chronic cutaneous lupus is selected from discoid lupus erythematosus (DLE), lupus erythematosus profundus (LEP), chilblain lupus erythematosus (CHLE), and lupus erythematosus tumidus (LET).
Discoid LE lesions are the most common lesions of Chronic cutaneous lupus erythematosus (CCLE) and appear as disk-shaped, round lesions. DLE occurs more frequently in women in their fourth and fifth decade of life. Patients with DLE generally have a more benign disease course as compared to patients with other CLE subtypes, with only most patients (-90-95%) never develop lupus in other organ systems (SLE) symptoms (see e.g. , Crowson & Magro, ./. Cutan. Pathol. 2001,
28(1): 1-23; and Chong et al., Br. J. Dermatol. 2012, 166(l):29-35). The sores usually appear on the scalp and face but sometimes they will occur on other parts of the body as well. Discoid lupus lesions are often red, scaly, and thick. Cisually, the lesions do not hurt or itch. Histologic examination of active DLE lesion reveals hyperkeratosis, dilated compact keratin-filled follicles, vacuolar degeneration of the basal keratinocytes, and an intensely inflammatory dermal infiltrate.
LE profundus (LEP), or panniculitis, features painful firm subcutaneous nodules occurring in areas of increased fat deposition, such as the upper arms and legs, face, and breasts. LEP tends to have a chronic course, characterized by remission and flares, and ultimately leaving atrophic scars (see e.g. , Fabbri et al., Am. J. Clin. Dermatol. 2003, 4(7):449-65). Histology shows lobular panniculitis with a dense lymphocytic infiltrate.
Chilblain lupus (CHLE) is a rare form of CCLE resembling frostbite. Lesions appear as painful, violaceous plaques and nodules in cold-exposed areas. Central erosions or ulcerations may occur on acral surfaces, such as fingers, toes, heels, nose, and ears. Chilblain lupus occurs when there is a temperature drop, and can be difficult to distinguish from frostbite. Pathology shows epidermal atrophy, interface vacuolization, and a perivascular mononuclear infiltrate. Approximately 80% of patients with CHLE will not develop SLE (see e.g., Hedrich et al., Clin. Rheumatol. 2008, 27(8):949-54).
Lupus tumidus is a subtype of CCLE characterized by extreme photosensitivity and a benign course occurring preferentially in men, unlike SLE which primarily occurs in women. Clinically, these lesions appear on the face as erythematous, edematous, urticaria-like polycylic plaques with sharp raised borders and smooth surfaces.
Primarily in young to middle aged women, subacute cutaneous lupus (SCLE) lesions are described as having a scaly red annular (“ring-like”) redness with central clearing. The lesions may also be polycyclic - that is, having the appearance of multiple rings coming together. There are two morphologic variants of SCLE: annular and papulosquamous. The annular variant is characterized by scaly annular erythematous plaques, which tend to coalesce and produce a polycyclic array (see e.g., Walling & Sontheimer, Am. J. Clin. Dermatol. 2009, 10(6):365-81). The papulosquamous variant can resemble eczema, as well as pityriasis in some instances (see e.g., Caproni et al. Int. J. Dermatol. 2001, 40(l):59-62). SCLE lesions occur in sun-exposed areas, including the upper thorax (‘V’ distribution), upper back, and the extensor surfaces of arms and forearms. About 10% of SLE patients will have SCLE lesions and those patients with systemic disease tend to have only mild symptoms in other organ compartments. Pathologic examination of SCLE lesions demonstrates hydropic degeneration of the basal keratinocytes, dermal edema, hyperkeratosis, follicular plugging, and a sparse superficial inflammatory infiltrate (see e.g, Fabbri et al , Am. J. Clin. Dermatol. 2003, 4(7):449-65).
First evidence for a functional role of type I IFNs in lupus erythematosus (i.e., LE) came from clinical observations in patients suffering from carcinoid tumors which were treated with recombinant IFNa and developed SLE due to this therapy (see e.g, Ronnblom et al, Acta Oncol. 1991, 30:537-540). This was supported by findings in multiple sclerosis patients who developed CLE-like lesions at the injection side of recombinant IFNp after subcutaneous application (see e.g, Arrue et al, J. Cutan. Pathol. 2007, 34, Suppl 1:18-21). Gene expression analyses in both SLE and CLE revealed a strong expression of type I IFN associated proinflammatory cytokines in blood and skin in association with disease activity (see e.g. , Kuhn et al, Semin. Immunopathol. 2016, 38:97-112; and Mikita et al, J. Dermatol. 2011, 38:839-849). These findings are supported by the data described herein, which demonstrates a strong expression of IFN-regulated genes and immune pathways within CLE skin lesions (see e.g. , FIGs. 1A-1D).
Previous reports describing strong IFN-signatures prompted the development of anti-IFN directed therapeutic strategies of LE patients. Anti-IFNa-agents (e.g, sifalimumab, rontalizumab) were the first drugs tested in clinical studies. These agents reduced the IFN signature in the blood but had limited effect on disease activity. Without being bound by theory, this was possibly due to the high variability of the different type I IFNs, including not only IFNa but also IFNP and IFNK binding to the same receptors (see e.g., Kalunian et al, Ann. Rheum. Dis. 2016, 75:196-202; Merrill et al, Ann. Rheum. Dis. 2011, 70:1905-1913; and Sarkar et al, Ann. Rheum. Dis. 2018, 77:1653-1664). Accordingly, targeting the common receptor was more effective: the anti-IFNa.p-receptor antibody anifrolumab significantly reduced the CLASI skin score in SLE patients in a recent clinical study (see e.g, Furie et al, Arthritis & Rheumatology, 2017, 69:376-386). Alternative strategies focused on “indirect” inhibition of the IFN system by (i) targeting the IFN-producing cells or (ii) the intracellular IFN-pathway. In skin and blood plasmacytoid DCs are regarded as the main IFN-producing cells (see e.g., Saadeh et al, Exp. Dermatol. 2016, 25:415- 421) and the pDC-specific antibodies BIIB059 and VIB7734 are now in clinical trials with first results indicating a decline of the CLASI-activity score (see e.g, Furie et al, Ann. Rheum. Dis. 2017, 76 (Suppl 2): 857).
The JAK family consists of four non-receptor tyrosine kinases (JAKl-3, TYK2) that transduce signals from growth factors and cytokines such as type I/III IFNs. JAK inhibitors were initially developed for the treatment of haematologic diseases with definite JAK-mutations showing anti clonal activity (see e.g, Morgan et al, Annu. Rev. Med. 2008, 59:213-222; and Pardanani et al, Leukemia 2008, 22:23- 30), but they also provide significant immunosuppressive effects (see e.g, Santos & Verstovsek, Expert Opin. Pharmacother. 2014, 15:1465-1473; Schwartz et al, Nat. Rev. Drug Discov. 2017, 16:843-862; Schwartz et al, Nat. Rev. Rheumatol. 2016, 12:25-36; and Howell et al, Ann. Allergy Asthma Immunol. 2018, 120:367-375).
Earlier studies revealed activation of JAK/STAT pathway and JAK protein expression in CLE and associated skin disorders (see e.g. , Scholtissek et al, J. Invest. Dermatol. 2017, 137:1484-1492; Alves de Medeiros et al, PLoS ONE 2016,
11 :e0164080; and Wenzel et al, J. Am. Acad. Dermatol. 2008, 58:437-442)). JAK inhibitors provided beneficial effects in interface dermatitides including graft-versus- host disease (see e.g., Spoerl et al, Blood , 2014, 123:3832-3842), dermatomyositis (see e.g., Hornung et al, N. Engl. J. Med. 2014, 371:2537-2538; and Homung et al, N. Engl. J. Med. 2015, 372:1274) and LE (see e.g., Wenzel et al, J. Invest. Dermatol. 2016, 136:1281-1283; and Briand et al, Ann. Rheum. Dis. 2019, 78:431-433) in addition to other inflammatory skin conditions (see e.g, Banerjee et al, Drugs, 2017, 77:521-546; Welsch et al, Eur. J. Immunol. 2017, 47:1096-1107; and Shreberk- Hassidim et al, J. Am. Acad. Dermatol. 2017, 76:745-753. el9). In CLE, JAK1/2 inhibitors ruxolitinib (see e.g., Wenzel et al, J. Invest. Dermatol. 2016, 136:1281- 1283; Briand et al, Ann. Rheum. Dis. 2019, 78:431-433; and Klaeschen et al, Exp. Dermatol. 2017, 26:728-730) and baricitinib (see e.g., Zimmerman et al, JAMA Dermatol. DOI: 10.1001/jamadermatol.2018.5077) as well as JAK1/3 inhibitor tofacitinib (see e.g., Konig et al, Ann. Rheum. Dis. 2017, 76:468-472) have been reported to be successful in patient’s treatment, and these drugs significantly decrease the expression of CLE-typical chemokines in vitro (see e.g, Klaeschen et al, Exp. Dermatol. 2017, 26:728-730; and Srivastava et al, Acta. Derm. Venereol. 2018, 98:772-775). One potential disadvantage of these drugs are side effects, including anemia and thrombopenia, which may occur in some patients. The side effects are associated with JAK2 and JAK3 inhibition.
Notably, recent reports of a double-blind placebo-controlled trial for the treatment of SLE suggest that baricitinib was not effective compared to placebo (see e.g., Wallace et al, Lancet, 2018, 392(10143):222-231; and Werth & Merrill, Br. J. Dermatol. 2019, 180(5): 964-965). Baricitinib (2mg or 4mg) was dosed once daily for 24 weeks. Eligible patients were aged 18 years or older, had a diagnosis of systemic lupus erythematosus (SLE), and had active disease involving skin or joints. The primary endpoint was the proportion of patients achieving resolution of arthritis or rash at week 24, as defined by Systemic Lupus Erythematosus Disease Activity Index-2000 (SLEDAI-2K).
In an exploratory end point, the Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) was evaluated, finding no improvement on the CLASI activity score with baricitinib. Unlike SLEDAI, the CLASI scoring system has been fully validated as a cutaneous- specific end point for SLE and successfully used in a number of phase II clinical trials for SLE and CLE. Recent reports also agree that the CLASI scoring system should be used as the skin outcome for lupus trials examining responsiveness in the skin (see e.s.. Albrecht J, et ah, J. Invest. Dermatol. 2005; 125:889-94; Klein et ah, Arch. Dermatol. 2011, 147:203-8; Furie et ah, Arthritis Rheumatol. 2017, 69:376-86; van Vollenhoven et ah, The Lancet 2018, 392:1330-9; Khamashta et ah, Ann. Rheumat. Dis. 2016, 75:1909-16; and Concha et ah, J. Invest. Dermatol. 2018, https://doi.org/10.1016/jjid.2018.08.017).
The methods provided herein utilize compounds or salts that are JAK1 inhibitors ( e.g ., JAKl selective inhibitors). In some embodiments, the compound is selected from:
{l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
4- { 3 -(Cyanomethyl)-3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl } -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
[3 -[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
4- [3 -(cy anomethyl)-3 -(3 ', 5 '-dimethyl - 1 H, 1 'H-4,4'-bipyrazol- 1 -yl)azetidin- 1 - yl]-2,5-difluoro-N-[(lS)-2,2,2-trifluoro-l-methylethyl]benzamide;
((2R,5S)-5-{2-[(lR)-l-hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin- 1 -yl }tetrahydro-2H-pyran-2-yl)acetonitrile;
3 -[ 1 -(6-chloropyridin-2-yl)pyrrolidin-3 -yl]-3 -[4-(7H-pyrrolo[2,3 -djpyrimidin- 4-yl)- lH-pyrazol- 1 -yljpropanenitrile;
3-(l-[l,3]oxazolo[5,4-b]pyridin-2-ylpyrrolidin-3-yl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]propanenitrile; 4-[(4- { 3 -cyano-2-[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]propyl}piperazin-l-yl)carbonyl]-3-fluorobenzonitrile;
4-[(4- { 3 -cyano-2-[3 -(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrrol- 1 - yl]propyl}piperazin-l-yl)carbonyl]-3-fluorobenzonitrile; [trans-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-3-(4-{[2-
(trifluoromethyl)pyrimidin-4-yl]carbonyl}piperazin-l-yl)cyclobutyl]acetonitrile;
{trans-3-(4-{[4-[(3-hydroxyazetidin-l-yl)methyl]-6-(trifluoromethyl)pyridin-
2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- y 1 ] cy cl obuty 1 } acetonitril e; {trans-3-(4-{[4-{[(2S)-2-(hydroxymethyl)pyrrolidin-l-yl]methyl}-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- lH-pyrazol- 1 -yljcyclobutyl } acetonitrile;
{trans-3-(4-{[4-{[(2R)-2-(hydroxymethyl)pyrrolidin-l-yl]methyl}-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile;
4-(4- { 3 - [(dimethyl amino)methyl] -5 -fluorophenoxy } piperidin- 1 -yl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]butanenitrile;
5-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl } -N-isopropylpyrazine-2-carboxamide; 4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl } -2,5-difluoro-N-[( 1 S)-2,2,2-trifluoro- 1 -methylethyljbenzamide;
5-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl } -N-isopropylpyrazine-2-carboxamide;
{l-(cis-4-{[6-(2-hydroxyethyl)-2-(trifluoromethyl)pyrimidin-4- yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-
3 -yl} acetonitrile;
{l-(cis-4-{[4-[(ethylamino)methyl]-6-(trifluoromethyl)pyridin-2- yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin- 3 -yl} acetonitrile; {l-(cis-4-{[4-(l -hydroxy- 1 -methyl ethyl)-6-(trifluoromethyl)pyri din-2- yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin- 3 -yl} acetonitrile; {l-(cis-4-{[4-{[(3R)-3-hydroxypyrrolidin-l-yl]methyl}-6-
(trifluoromethyl)pyridin-2-yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyriinidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{ 1 -(ci s-4- { [4- { [(3 S)-3 -hy droxypyrrolidin- 1 -yljmethyl } -6- (trifluoromethyl)pyridin-2-yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyriinidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{trans-3-(4-{[4-({[(lS)-2-hydroxy-l-methylethyl]amino}methyl)-6- (trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)- lH-pyrazol- 1 -yljcyclobutyl } acetonitrile;
{trans-3-(4-{[4-({[(2R)-2-hydroxypropyl]amino}methyl)-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-
4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile;
{trans-3-(4-{[4-({[(2S)-2-hydroxypropyl]amino}methyl)-6- (trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile; and
{trans-3 -(4- { [4-(2 -hydroxy ethyl)-6-(trifluoromethyl)pyridin-2- yl]oxy }piperidin- 1 -yl)- 1 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 - y 1 ] cy cl obuty 1 } acetonitril e; or a pharmaceutically acceptable salt of any of the aforementioned.
In some embodiments, the compounds or salt is selective for JAK1 over JAK2, JAK3 and TYK2. For example, some of the compounds described herein, or a pharmaceutically acceptable salts thereof, preferentially inhibit JAK1 over one or more of JAK2, JAK3, and TYK2. JAK1 plays a central role in a number of cytokine and growth factor signaling pathways that, when dysregulated, can result in or contribute to disease states. For example, IL-6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested to have detrimental effects (see e.g ., Fonesca, et al., Autoimmunity Reviews, 8:538-42, 2009). Because IL-6 signals, at least in part, through JAK1, IL-6 can be indirectly through JAK1 inhibition, resulting in potential clinical benefit (see e.g, Guschin, et al. Embo J 14:1421, 1995; and Smolen, et al. Lancet 371:987, 2008). In other autoimmune diseases and cancers, elevated systemic levels of inflammatory cytokines that activate JAK1 may also contribute to the disease and/or associated symptoms. Therefore, patients with such diseases may benefit from JAK1 inhibition. Selective inhibitors of JAK1 may be efficacious while avoiding unnecessary and potentially undesirable effects of inhibiting other JAK kinases, as described herein.
In some embodiments, the compounds or salt inhibits JAK1 preferentially over JAK2 (e.g, having a JAK2/JAK1 IC50 ratio >1). In some embodiments, the compounds or salts provided herein are about 10-fold more selective for JAK1 over JAK2. In some embodiments, the compounds or salts provided herein are about 3- fold, about 5-fold, about 10-fold, about 15-fold, or about 20-fold more selective for JAK1 over JAK2 as calculated by measuring IC50 at 1 mM ATP (see Example 1). In some embodiments, the JAK1 inhibitor is a compound of Table 1, or a pharmaceutically acceptable salt thereof. The compounds in Table 1 are JAK1 selective inhibitors (e.g, selective over JAK2, JAK3, and TYK2). The IC50 values obtained by the method of Example 1 at 1 mM ATP are shown in Table 1.
Table 1.
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
+ means <10 nM (see Example 1 for assay conditions)
++ means < 100 nM (see Example 1 for assay conditions) +++ means < 300 nM (see Example 1 for assay conditions) aData for enantiomer 1 bData for enantiomer 2
In some embodiments, the JAK1 inhibitor is {l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 inhibitor is (l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt.
The synthesis and preparation of (l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile and the adipic acid salt of the same can be found, e.g., in US Patent Publ. No. 2011/0224190, filed March 9, 2011, US Patent Publ. No. 2013/0060026, filed September 6, 2012, and US Patent Publ. No. 2014/0256941, filed March 5, 2014, each of which is incorporated herein by reference in its entirety. In some embodiments, the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide, or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide phosphoric acid salt.
In some embodiment, the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide hydrochloric acid salt.
In some embodiment, the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide hydrobromic acid salt.
In some embodiment, the JAK1 inhibitor is 4-[3-(cyanomethyl)-3-(3',5'- dimethyl-lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide sulfuric acid salt.
The synthesis and preparation of 4-[3-(cyanomethyl)-3-(3',5'-dimethyl- lH,m-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2-trifluoro-l- methylethyljbenzamide and the phosphoric acid salt of the same can be found, e.g., in US Patent Publ. No. US 2014/0343030, filed May 16, 2014, which is incorporated herein by reference in its entirety.
In some embodiments, the JAK1 inhibitor is ((2R,5S)-5-{2-[(lR)-l- hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl}tetrahydro-2H-pyran-2- yl)acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 inhibitor is ((2R,5S)-5-{2-[(lR)-l- hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl}tetrahydro-2H-pyran-2- yl)acetonitrile monohydrate.
Synthesis of ((2R,5S)-5-{2-[(lR)-l-hydroxyethyl]-lH-imidazo[4,5- d]thieno[3,2-b]pyridin-l-yl}tetrahydro-2H-pyran-2-yl)acetonitrile and characterization of the anhydrous and monohydrate forms of the same are described in US Patent Publ. No. 2014/0121198, filed October 31, 2013 and US Patent Publ. No. 2015/0344497, filed April 29, 2015, each of which is incorporated herein by reference in its entirety. In some embodiments, the compounds of Table 1 are prepared by the synthetic procedures described in US Patent Publ. No. 2011/0224190, filed March 9, 2011, US Patent Publ. No. 2014/0343030, filed May 16, 2014, US Patent Publ. No. 2014/0121198, filed October 31, 2013, US Patent Publ. No. 2010/0298334, filed May 21, 2010, US Patent Publ. No. 2011/0059951, filed August 31, 2010, US Patent Publ.
No. 2012/0149681, filed November 18, 2011, US Patent Publ. No. 2012/0149682, filed November 18, 2011, US Patent Publ. 2013/0018034, filed June 19, 2012, US Patent Publ. No. 2013/0045963, filed August 17, 2012, and US Patent Publ. No. 2014/0005166, filed May 17, 2013, each of which is incorporated herein by reference in its entirety.
In some embodiments, the JAK1 inhibitor is selected from the compounds, or pharmaceutically acceptable salts thereof, of US Patent Publ. No. 2011/0224190, filed March 9, 2011, US Patent Publ. No. 2014/0343030, filed May 16, 2014, US Patent Publ. No. 2014/0121198, filed October 31, 2013, US Patent Publ. No. 2010/0298334, filed May 21, 2010, US Patent Publ. No. 2011/0059951, filed August 31, 2010, US Patent Publ. No. 2012/0149681, filed November 18, 2011, US Patent Publ. No. 2012/0149682, filed November 18, 2011, US Patent Publ. 2013/0018034, filed June 19, 2012, US Patent Publ. No. 2013/0045963, filed August 17, 2012, and US Patent Publ. No. 2014/0005166, filed May 17, 2013, each of which is incorporated herein by reference in its entirety.
In some embodiments, the JAK1 inhibitor is a compound of Formula I:
Figure imgf000024_0001
or a pharmaceutically acceptable salt thereof, wherein: X is N or CH;
L is C(=0) or C(=0)NH;
A is phenyl, pyridinyl, or pyrimidinyl each of which is optionally substituted with 1 or 2 independently selected R1 groups; and each R1 is, independently, fluoro, or trifluoromethyl.
In some embodiments, the compound of Formula I is {l-{l-[3-fluoro-2- (trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)- lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of Formula I is 4-{3-(Cyanomethyl)-3- [4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 -yljazetidin- 1 -yl } -N-[4-fluoro-2- (trifluoromethyl)phenyl]piperidine-l -carboxamide, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of Formula I is [3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)- lH-pyrazol- 1 -yl]- 1 -( 1 - { [2-(trifluoromethyl)pyrimidin-4- yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 inhibitor is a compound of Formula II:
Figure imgf000025_0001
II or a pharmaceutically acceptable salt thereof, wherein:
R2 is Ci-6 alkyl, Ci-6 haloalkyl, C3-6 cycloalkyl, or C3-6 cycloalkyl-Ci-3 alkyl, wherein said Ci-6 alkyl, C3-6 cycloalkyl, and C3-6 cycloalkyl-Ci-3 alkyl, are each optionally substituted with 1, 2, or 3 substituents independently selected from fluoro, - CF3, and methyl;
R3 is H or methyl; R4 is H, F, or Cl; R5 is H or F; R6 is H or F;
R7 is H or F;
R8 is H or methyl;
R9 is H or methyl;
R10 is H or methyl; and R11 is H or methyl.
In some embodiments, the compound of Formula II is 4-[3-(cyanomethyl)-3- (3',5'-dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5-difluoro-N-[(lS)-2,2,2- trifluoro-l-methylethyl]benzamide , or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 inhibitor is a compound of Formula III:
Figure imgf000026_0001
or a pharmaceutically acceptable salt thereof, wherein:
Cy4 is a tetrahydro-2H-pyran ring, which is optionally substituted with 1 or 2 groups independently selected from CN, OH, F, Cl, C1-3 alkyl, C1-3 haloalkyl, CN-C1-3 alkyl, HO-C1-3 alkyl, amino, C1-3 alkylamino, and di(Ci-3 alkyl)amino, wherein said Ci-3 alkyl and di(Ci-3 alkyl)amino is optionally substituted with 1, 2, or 3 substituents independently selected from F, Cl, C1-3 alkylaminosulfonyl, and C1-3 alkyl sulfonyl; and
R12 is -CH2-OH, -CH(CH3)-OH, or -CH2-NHSO2CH3.
In some embodiments, the compound of Formula III is ((2R,5S)-5-{2-[(lR)-l- hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl}tetrahydro-2H-pyran-2- yl)acetonitrile, or a pharmaceutically acceptable salt thereof.
One or more constituent atoms of the compounds described herein can be replaced or substituted with isotopes of the atoms in natural or non-natural abundance. In some embodiments, the compound includes at least one deuterium atom. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1-2, 1-3, 1-4, 1-5, or 1-6 deuterium atoms. In some embodiments, all of the hydrogen atoms in a compound can be replaced or substituted by deuterium atoms.
Synthetic methods for including isotopes into organic compounds are known in the art (Deuterium Labeling in Organic Chemistry by Alan F. Thomas (New York, N.Y., Appleton-Century-Crofts, 1971; The Renaissance of H/D Exchange by Jens Atzrodt, Volker Derdau, Thorsten Fey and Jochen Zimmermann, Angew. Chem. Int. Ed. 2007, 7744-7765; The Organic Chemistry of Isotopic Labelling by James R. Hanson, Royal Society of Chemistry, 2011). Isotopically labeled compounds can be used in various studies such as NMR spectroscopy, metabolism experiments, and/or assays.
Substitution with heavier isotopes, such as deuterium, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances (see e.g ., A. Kerekes et. al. J. Med. Chem. 2011, 54, 201-210; R. Xu et. al. J. Label Compd. Radiopharm. 2015, 58, 308-312). In particular, substitution at one or more metabolism sites may afford one or more of the therapeutic advantages.
Accordingly, in some embodiments, the JAK1 inhibitor (e.g. , the JAK1 selective inhibitor) is a compound, wherein one or more hydrogen atoms in the compound are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof.
As used herein, the phrase “optionally substituted” means unsubstituted or substituted. As used herein, the term “substituted” means that a hydrogen atom is removed and replaced by a substituent. It is to be understood that substitution at a given atom is limited by valency.
As used herein, the term “Cn-m alkyl”, employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbon atoms. In some embodiments, the alkyl group contains 1 to 6, or 1 to 3 carbon atoms. Examples of alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, /e/V-butyl , «-pentyl, 2-methyl-l-butyl, 3-pentyl, «-hexyl, 1,2,2- trimethylpropyl, and the like. As used herein, the term “alkylene”, employed alone or in combination with other terms, refers to a divalent alkyl linking group, which can be branched or straight-chain, where the two substituents may be attached any position of the alkylene linking group. Examples of alkylene groups include, but are not limited to, ethan-l,2-diyl, propan- 1, 3 -diyl, propan- 1,2-diyl, and the like.
As used herein, the term “HO-Ci-3-alkyl” refers to a group of formula - alkylene-OH, wherein said alkylene group has 1 to 3 carbon atoms.
As used herein, the term “CN-C1-3 alkyl” refers to a C1-3 alkyl substituted by a cyano group.
As used herein, the term “amino” refers to a group of formula -NH2.
As used herein, the term “di(Ci-3-alkyl)amino” refers to a group of formula - N(alkyl)2, wherein the two alkyl groups each has, independently, 1 to 3 carbon atoms.
As used herein, the term “C1-3 alkylamino” refers to a group of formula -NH(alkyl), wherein the alkyl group has 1 to 3 carbon atoms.
As used herein, the term “di(Ci-3 alkyl)aminosulfonyl” refers to a group of formula -S(0)2N(alkyl)2, wherein each alkyl group independently has 1 to 3 carbon atoms.
As used herein, the term “C1-3 alkyl sulfonyl” refers to a group of formula -S(0)2-alkyl, wherein the alkyl group has 1 to 3 carbon atoms.
As used herein, “halo” or “halogen”, employed alone or in combination with other terms, includes fluoro, chloro, bromo, and iodo. In some embodiments, the halo group is fluoro or chloro.
As used herein, the term “Cn-mhaloalkyl”, employed alone or in combination with other terms, refers to a Cn-m alkyl group having up to {2(n to m)+l } halogen atoms which may either be the same or different. In some embodiments, the halogen atoms are fluoro atoms. In some embodiments, the alkyl group has 1-6 or 1-3 carbon atoms. Example haloalkyl groups include CF3, C2F5, CHF2, CCh, CHCI2, C2CI5, and the like. In some embodiments, the haloalkyl group is a fluoroalkyl group.
As used herein, the term “C1-3 fluoroalkyl” refers to a C1-3 alkyl group that may be partially or completely substituted by fluoro atoms.
As used herein, the term “C3-6 cycloalkyl”, employed alone or in combination with other terms, refers to a non-aromatic monocyclic hydrocarbon moiety, having 3- 6 carbon atoms, which may optionally contain one or more alkenylene groups as part of the ring structure. One or more ring-forming carbon atoms of a cycloalkyl group can be oxidized to form carbonyl linkages. Exemplary C3-6 cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, and the like. In some embodiments, the cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
As used herein, the term “C3-6 cycloalkyl-Ci-3 alkyl” refers to a group of formula -C1-3 alkylene- C3-6 cycloalkyl.
The compounds described herein can be asymmetric ( e.g ., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present application. Cis and trans geometric isomers of the compounds of the present application are described and may be isolated as a mixture of isomers or as separated isomeric forms. In some embodiments, the compound has the (^-configuration. In some embodiments, the compound has the (Sj- configuration.
Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallization using a chiral resolving acid which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as b-camphorsulfonic acid. Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a-methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N- methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, and the like. Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent ( e.g ., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
Compounds described herein include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. For example, it will be recognized that the following pyrazole ring may form two tautomers:
Figure imgf000030_0001
It is intended that the claims cover both tautomers.
All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g. hydrates and solvates) or can be isolated.
In some embodiments, the compounds described herein, or salts thereof, are substantially isolated. By “substantially isolated” is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compounds described herein. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds described herein, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The expressions, “ambient temperature” and “room temperature” or “rt” as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20 °C to about 30 °C.
The present application also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present application include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present application can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences , 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science , 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
As used herein, the term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, “contacting” a JAK with a compound of the invention includes the administration of a compound of the present application to an individual or patient, such as a human, having a JAK, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the JAK. As used herein, the term “subject”, “individual” or “patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans. In some embodiments, the “subject,” “individual,” or “patient” is in need of said treatment.
In some embodiments, the inhibitors are administered in a therapeutically effective amount. As used herein, the phrase “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician. .
As used herein, the term “treating” or “treatment” refers to one or more of (1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); (2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease; or (3) preventing the disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease. In some embodiments, treating refers to inhibiting or ameliorating the disease. In some embodiments, treating is preventing the disease.
Combination Therapies
The methods described herein can further comprise administering one or more additional therapeutic agents. The one or more additional therapeutic agents can be administered to a patient simultaneously or sequentially.
In some embodiments, the additional therapeutic agent is an antibiotic. In some embodiments, the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, erythromycin, metronidazole, rifampin, moxifloxacin, dapsone, or a combination thereof. In some embodiments, the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, or erythromycin in combination with metronidazole. In some embodiments, the antibiotic is a combination of rifampin, moxifloxacin, and metronidazole. In some embodiments, the antibiotic is a combination of moxifloxacin and rifampin.
In some embodiments, the additional therapeutic agent is a retinoid. In some embodiments, the retinoid is etretinate, acitretin, or isotretinoin.
In some embodiments, the additional therapeutic agent is a steroid. In some embodiments, the additional therapeutic agent is a corticosteroid. In some embodiments, the steroid is such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisone, prednisolone, or flumetholone.
In some embodiments, the additional therapeutic agent is an anti-TNF-alpha agent. In some embodiments, the anti-TNF-alpha agent is an anti-TNF-alpha antibody. In some embodiments, the anti-TNF-alpha agent is infliximab or etanercept, or adalimumab.
In some embodiments, the additional therapeutic agent is an immunosuppressant. In some embodiments, the immunosuppressant is methotrexate or cyclosporin A. In some embodiments, the immunosuppressant is mycophenolate mofetil or mycophenolate sodium.
In some embodiments, the additional therapeutic agent is finasteride, metformin, adapalene or azelaic acid.
In some embodiments, the method further comprises administering an additional therapeutic agent selected from IMiDs, an anti-IL-6 agent, a hypomethylating agent, and a biologic response modifier (BRM).
Generally, a BRM is a substances made from living organisms to treat disease, which may occur naturally in the body or may be made in the laboratory. Examples of BRMs include IL-2, interferon, various types of colony-stimulating factors (CSF, GM-CSF, G-CSF), monoclonal antibodies such as abciximab, etanercept, infliximab, rituximab, trasturzumab, and high dose ascorbate.
In some embodiments, the hypomethylating agent is a DNA methyltransferase inhibitor. In some embodiments, the DNA methyltransferase inhibitor is selected from 5 azacytidine and decitabine. Generally, IMiDs are as immunomodulatory agents. In some embodiments, the IMiD is selected from thalidomide, lenalidomide, pomalidomide, CC-11006, and CC-10015.
In some embodiments, the method further comprises administering an additional therapeutic agent selected from anti -thymocyte globulin, recombinant human granulocyte colony-stimulating factor (G CSF), granulocyte-monocyte CSF (GM-CSF), an erythropoiesis-stimulating agent (ESA), and cyclosporine.
In some embodiments, the method further comprises administering an additional JAK inhibitor to the patient. In some embodiments, the additional JAK inhibitor is selected from a JAK2 inhibitor (e.g, a selective JAK2 inhibitor), a JAK1/JAK2 inhibitor, a JAK3 inhibitor (e.g, a selective JAK3 inhibitor), and a JAK1/JAK3 inhibitor, or a pharmaceutically acceptable salt of any of the aforementioned.
In some embodiments, the additional JAK inhibitor is a JAK1/JAK2 inhibitor. In some embodiments, the JAK1/JAK2 inhibitor is selective for JAK1 and JAK2 over JAK3 and TYK2. In some embodiments, the JAK1/JAK2 inhibitor is 3-cyclopentyl-3- [4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]propanenitrile, or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK2 inhibitor is (3R)-3-cyclopentyl-3-[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljpropanenitrile (ruxolitinib), or a pharmaceutically acceptable salt thereof. Ruxolitinib has an ICso of less than 10 nM at 1 mM ATP at JAK1 and JAK2. 3- Cyclopentyl-3-[4-(7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]propanenitrile and ruxolitinib can be made by the procedure described in US 7,598,257 (Example 67), filed December 12, 2006, which is incorporated herein by reference in its entirety. In some embodiments, the JAK1/JAK2 inhibitor is (3R)-3-cyclopentyl-3-[4- (7H-pynOlo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]propanenitrile phosphoric acid salt. The phosphoric acid salt can be made as described in U.S. Patent 8,722,693, which is incorporated herein by reference in its entirety. In some embodiments, the JAK1/JAK2 inhibitor is barcitinib, or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK2 inhibitor is barcitinib.
In some embodiments, the additional JAK1 inhibitor is a JAK1/JAK3 inhibitor. In some embodiments, the JAK1/JAK3 inhibitor is tofacitinib, or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK3 inhibitor is tofacitinib.
In some embodiments, the JAK1/JAK2 inhibitor can be an isotopically-labeled compound, or a pharmaceutically acceptable salt thereof. An “isotopically” or “radio- labeled” compound is a compound wherein one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present disclosure include but are not limited to 2H (also written as D for deuterium), 3H (also written as T for tritium), UC, 13C, 14C, 13N, 15N, 150, 170, 180, 18F, 35 S, 36C1, 82Br, 75Br, 76Br, 77Br, 123I, 124I, 125I and 131I. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced by deuterium atoms, such as -CD3 being substituted for -CFb).
Accordingly, in some embodiments, the JAK1/JAK2 inhibitor is a compound, wherein one or more hydrogen atoms in the compound are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1/JAK2 inhibitor is ruxolitinib, wherein one or more hydrogen atoms are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK2 inhibitor is any of the compounds in US Patent 9,249,149, which is incorporated herein by reference in its entirety, or a pharmaceutically acceptable salt thereof. In some embodiments, the inhibitor of JAK1/JAK2 is CTP-543, or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1/JAK2 inhibitor is a compound of Formula IV:
Figure imgf000036_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is selected from H and D; each R2 is independently selected from H and D, provided that each R2 attached to a common carbon is the same; each R3 is independently selected from H and D, provided that each R3 attached to a common carbon is the same;
R4 is selected from H and D; each R5 is the same and is selected from H and D; and
R6, R7, and R8 are each independently selected from H and D; provided that when R1 is H, each R2 and each R3 are H, R4 is H, and each of R6, R7, and R8 is H, then each R5 is D.
In some embodiments, the JAK1/JAK2 inhibitor is a compound of Formula IV selected from the following compounds 100-130 in the table below (wherein R6, R7, and R8 are each H), or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK2 inhibitor is a compound of Formula IV selected from the following compounds 200-231 in the table below (wherein R6, R7, and R8 are each D), or a pharmaceutically acceptable salt thereof.
Figure imgf000037_0001
Figure imgf000038_0001
In some embodiments, the JAK1/JAK2 inhibitor is baricitinib, wherein one or more hydrogen atoms are replaced by deuterium atoms, or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1/JAK2 inhibitor is any of the compounds in US Patent 9,540,367 (which is incorporated herein by reference in its entirety), or a pharmaceutically acceptable salt thereof.
In some embodiments, the additional JAK inhibitor is selected from baricitinib, tofacitinib, oclacitinib, filgotinib, gandotinib, lestaurtinib, momelotinib, bacritinib, PF-04965842, upadacitinib, peficitinib, fedratinib, cucurbitacin I, ATI-501 (Aclaris), ATI-502 (Aclaris), JTE-052 (z.e., delgocitinib; Leo Pharma and Japan Tobacco), and CHZ868.
Additional agents including, but not limited to, anti-inflammatory agents, immunosuppressants, PI3K6 inhibitors, mTor inhibitors, Bcr-Abl inhibitors, Flt-3 inhibitors, RAF inhibitors, and FAK kinase inhibitors ( e.g ., those described in WO 2006/056399, which is incorporated herein by reference in its entirety), or other agents can be used in combination with the JAK1 inhibitors described herein for treatment of cutaneous lupus erythematosus (CLE). The one or more additional agents can be administered to a patient simultaneously or sequentially.
Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No.
5,521,184, WO 04/005281, and U.S. Ser. No. 60/578,491, all of which are incorporated herein by reference in their entirety.
Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120, all of which are incorporated herein by reference in their entirety.
Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444, both of which are incorporated herein by reference in their entirety.
Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402, all of which are incorporated herein by reference in their entirety.
In some embodiments, one or more of the JAK1 inhibitors described herein can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
In some embodiments, the additional therapeutic agent is fluocinolone acetonide (Retisert®) or rimexolone (AL-2178, Vexol, Alcon).
In some embodiments, the additional therapeutic agent is cyclosporine (Restasis®). In some embodiments, the additional therapeutic agent is selected from Dehydrex™ (Holies Labs), Civamide (Opko), sodium hyaluronate (Vismed, Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine, doxycycline (ALTY-0501, Alacrity), minocycline, iDestrin™ (NP50301, Nascent Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline (Duramycin, MOLI1901, Lantibio), CF101 (2S,3S,4R,5R)-3,4-dihydroxy-5-[6-[(3- iodophenyl)methylamino]purin-9-yl]-N-methyl-oxolane-2-carbamyl, Can-Fite Biopharma), voclosporin (LX212 or LX214, Lux Biosciences), ARG103 (Agentis), RX- 10045 (synthetic resolvin analog, Resolvyx), DYN15 (Dyanmis Therapeutics), rivoglitazone (DE011, Daiichi Sanko), TB4 (RegeneRx), OPH-01 (Ophtalmis Monaco), PCS101 (Pericor Science), REVl-31 (Evolutec), Lacritin (Senju), rebamipide (Otsuka-Novartis), OT-551 (Othera), PAI-2 (University of Pennsylvania and Temple University), pilocarpine, tacrolimus, pimecrolimus (AMS981, Novartis), loteprednol etabonate, rituximab, diquafosol tetrasodium (INS365, Inspire), KLS- 0611 (Kissei Pharmaceuticals), dehydroepiandrosterone, anakinra, efalizumab, mycophenolate sodium, etanercept (Embrel®), hydroxychloroquine, NGX267 (TorreyPines Therapeutics), actemra, gemcitabine, oxaliplatin, L-asparaginase, or thalidomide.
In some embodiments, the additional therapeutic agent is an anti -angiogenic agent, cholinergic agonist, TRP-1 receptor modulator, a calcium channel blocker, a mucin secretagogue, MUC1 stimulant, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, or an mTOR inhibitor.In some embodiments, the additional therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycline). In some embodiments, the additional therapeutic agent binds to FKBP12.
In some embodiments, the additional therapeutic agent is an alkylating agent or DNA cross-linking agent; an anti-metabolite/demethylating agent (e.g., 5- flurouracil, capecitabine or azacitidine); an anti -hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e.g. vincristine or paclitaxel); an topoisomerase (I or II) inhibitor (e.g. mitoxantrone and irinotecan); an apoptotic inducers (e.g. ABT-737); a nucleic acid therapy (e.g. antisense or RNAi); nuclear receptor ligands (e.g., agonists and/or antagonists: all- trans retinoic acid or bexarotene); epigenetic targeting agents such as histone deacetylase inhibitors (e.g. vorinostat), hypomethylating agents (e.g. decitabine); regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
In some embodiments, the additional therapeutic agent is selected from an antibiotic, an antiviral, an antifungal, an anesthetic, an anti-inflammatory agent (e.g., steroidal and non-steroidal anti-inflammatory agents), and an anti-allergic agent. Examples of suitable agents include, but are not limited to, aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine; sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin; colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives (“rifampins”); cycloserine; beta-lactams; cephalosporins; amphotericins; fluconazole; flucytosine; natamycin; miconazole; ketoconazole; corticosteroids; diclofenac; flurbiprofen; ketorolac; suprofen; cromolyn; lodoxamide; levocabastin; naphazoline; antazoline; pheniramine; or azalide antibiotic.
Pharmaceutical Formulations and Dosage Forms
When employed as pharmaceuticals, the JAK1 inhibitors provided herein can be administered in the form of pharmaceutical compositions. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including intradermal, transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g, by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g, intrathecal or intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, dermal patches, solutions, suspensions, foams, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids, and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. In some embodiments, the composition is formulated for topical administration by transdermal patch, dermal patch, solution, suspension, gel, cream, ointment, lotion, spray, foam, liquid, drops, suppository, and powder. In some embodiments, the composition is formulated for topical administration by transdermal patch. In some embodiments, the composition is formulated for topical administration by dermal patch. In some embodiments, the composition is formulated as a foam ( e.g ., for topical administration).
For the treatment of skin disorders such as cutaneous lupus erythematosus (CLE), topical drugs which are able to penetrate the skin barrier and provide limited systemic effects are of particular importance.
Topical (dermal/intradermal) formulations are typically solutions, suspensions, gels, creams, ointments, lotions, sprays and foams. Preferred topical formulations should be physically and chemically stable, not cause skin irritation, and deliver the active agent (e.g., a selective JAK1 inhibitor, or a pharmaceutically acceptable salt thereof, as described herein) at the appropriate layer of the skin in concentrations that would result in therapeutic response, with limited systemic exposure.
In some embodiments, the administration is topical and comprised of formulations with one or more pharmaceutically (e.g, dermatologically) acceptable excipients. Examples of dermatologically acceptable excipients include, but are not limited to, a pH adjusting agents, chelating agents, preservatives, co-solvents, penetration enhancers, humectants, thickening, gelling, viscosity building agents, surfactants, propellants, fragrance, colorants, or any combination or mixture thereof.
In some embodiments, the topical formulation is administered locally to the patient (e.g, administered at the site of a lesion).
In some embodiments, the pH-adjusting agent is selected from an acid, an acid salt, a base, a base salt, and a buffer, or any mixture thereof. Exemplary acids include, but are not limited to, lactic acid, acetic acid, citric acid, and benzoic acid, and salts thereof. Exemplary bases include, but are not limited to, trolamine, tromethamine, and salts thereof. Exemplary buffers include, but are not limited to, citrate/citric acid, acetate/acetic acid, edetate/edetic acid, lactate/lactic acid, and the like.
In some embodiments, the chelating agent is a single excipient. In some embodiments, the chelating agent is a mixture of two or more chelating agents. Exemplary chelating agents include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), or a salt thereof. In some embodiments, the chelating agent comprises a mixture of a chelating agent and an antioxidant, wherein the chelating agent and antioxidant prevent, minimize, or reduce oxidative degradation reactions in the composition. Exemplary anti-oxidants include, but are not limited to, butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), tocopherol, and propyl gallate.
In some embodiments, the composition comprises one or more preservatives. In some embodiments, the composition comprises a mixture of two or more preservatives. In some embodiments, the composition comprises one to five preservatives. Exemplary preservatives include, but are not limited to, benzyl alcohol, phenonyexthanol, methyl paraben, ethyl paraben, propyl paraben, butyl paraben, and imidazolidinyl urea.
In some embodiments, the composition comprises one or more co-solvents. In some embodiments, the composition comprises a mixture of two or more co-solvents. In some embodiments, the composition comprises one to five co-solvents. Exemplary solvents include, but are not limited to, water, propylene glycol, diethylene glycol monoethyl ether, dimethyl isosorbide, ethyl alcohol, isopropyl alcohol, benzyl alcohol, propanediol, propylene glycol, polyethylene glycols ( e.g ., polyethylene glycol 200, polyethylene glycol 300, polyethylene glycol 400, and the like). In some embodiments, the solvent is a non-water soluble agent. Exemplary non-water soluble agents include, but are not limited to, diethyl sebacate, diisopropyl adipate, isopropyl myristate, isopropyl palmitate, and medium chain triglycerides.
In some embodiments, the composition comprises one or more penetration enhancers. In some embodiments, the composition comprises a mixture of two or more penetration enhancers. In some embodiments, the composition comprises one to five penetration enhancers. The penetration enhancers can act as both a solvent and a penetration enhancer. Exemplary penetration enhancers include, but are not limited to, fatty acids, fatty acid esters, fatty alcohols, pyrrolidones, sulfoxides, alcohols, diols and polyols, or any mixture thereof. In some embodiments, a co-solvent provided herein is a penetration enhancer.
In some embodiments, the composition comprises one or more thickening, gelling, or viscosity building agents. In some embodiments, the composition comprises a mixture of two or more thickening, gelling, or viscosity building agents. In some embodiments, the composition comprises one to five thickening, gelling, or viscosity building agents. Exemplary thickening, gelling, or viscosity building agents include, but are not limited to, cellulosic derivatives ( e.g ., hydroxy ethylcellulose (HEC), carboxymethylcellulose, hydroxypropylcellulose (HPC), and hydroxypropyl methylcellulose (HPMC), and polyvinylpyrrolidone (PVP).
The surfactant is a compound that lowers the surface tension between two liquids or between a liquid and a solid. Surfactant may be a mixture of two or more surfactants. Exemplary surfactants include, but are not limited to, ethoxylated fatty alcohol ether (e.g., steareth-2, steareth-10, steareth-20, ceteareth-2, ceteareth-10, and the like), PEG esters (e.g., PEG-4 dilaurate, PEG-20 stearate, and the like), Glyceryl esters or derivatives thereof (e.g, glyceryl dioleate, glyceryl stearate, and the like), polymeric ethers (e.g, poloxamer 124, poloxamer 181, poloxamer 182, and the like), sorbitan derivatives (e.g, polysorbate 80, sorbitan monostearate, and the like), fatty alcohols (e.g, cetyl alcohol, stearyl alcohol, cetearyl alcohol, and the like), and emulsifying wax (e.g, emulsifying wax NF, mixtures of mixture of cetearyl alcohol and polysorbate 60, and the like).
In some embodiments, the administration is topical administration to the skin.
In some embodiments, the administration is oral.
In some embodiments, administration of a topical composition comprising a selective JAK1 inhibitor provided herein to a patient provides a systemic exposure (Cmax) at steady state of about 5% or less of the applied dose, e.g, about 4% or less, about 3% or less, about 2% or less, about 1% or less, and the like. In some embodiments, administration of a topical composition comprising a selective JAK1 inhibitor provided herein to a patient provides a systemic exposure (Cmax) at steady state of about 1% or less of the applied dose, This invention also includes pharmaceutical compositions which contain, as the active ingredient, a JAK1 inhibitors provided herein, or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients). In some embodiments, the composition is suitable for topical administration. In making the compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
The JAK1 inhibitors provided herein may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. Finely divided (nanoparticulate) preparations of the JAK1 inhibitors can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
In some embodiments, the pharmaceutical composition comprises silicified microcrystalline cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the silicified microcrystalline cellulose comprises about 98% microcrystalline cellulose and about 2% silicon dioxide w/w.
In some embodiments, the composition is a sustained release composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and at least one component selected from microcrystalline cellulose, lactose monohydrate, hydroxypropyl methylcellulose, and polyethylene oxide. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate, and hydroxypropyl methylcellulose. In some embodiments, the composition comprises at least one compound described herein, or a pharmaceutically acceptable salt thereof, and microcrystalline cellulose, lactose monohydrate, and polyethylene oxide. In some embodiments, the composition further comprises magnesium stearate or silicon dioxide. In some embodiments, the microcrystalline cellulose is Avicel PHI 02™. In some embodiments, the lactose monohydrate is Fast-flo 316™. In some embodiments, the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M Premier™) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel K00LV™). In some embodiments, the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Poly ox WSR 1105™).
In some embodiments, a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.
The compositions can be formulated in a unit dosage form, each dosage containing from about 1 to about 1,000 mg, from about 1 mg to about 100 mg, from 1 mg to about 50 mg, and from about 1 mg to 10 mg of active ingredient. Preferably, the dosage is from about 1 mg to about 50 mg or about 1 mg to about 10 mg of active ingredient. In some embodiments, each dosage contains about 10 mg of the active ingredient. In some embodiments, each dosage contains about 50 mg of the active ingredient. In some embodiments, each dosage contains about 25 mg of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
In some embodiments, the compositions comprise from about 1 to about 1,000 mg, from about 1 mg to about 100 mg, from 1 mg to about 50 mg, and from about 1 mg to 10 mg of active ingredient. Preferably, the compositions comprise from about 1 mg to about 50 mg or about 1 mg to about 10 mg of active ingredient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 1 mg to about 10 mg, about 1 mg to about 20 mg, about 1 mg to about 25 mg, about 1 mg to about 50 mg of the active ingredient.
The active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present application. When referring to these preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present application (e.g. a JAK1 inhibitor provided herein).
The tablets or pills of the present application can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
The liquid forms in which the compounds and compositions of the present application can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
Topical (e.g, intradermal) administration provides the advantage of treating the skin disorder locally, minimizing potential adverse events associated with systemic exposure, and allowing an easier discontinuation of the therapy, if necessary. Additionally, some topical dosage forms such as creams, ointments, and gels have the benefit of excipients that may act as emollients or occlusive agents, which can increase patient well-being and compliance during the treatment period. Other dosage routes such as oral, parenteral, and inhalation may lead to supratherapeutic systemic drug levels, increased likelihood of adverse events, drug-drug interactions, and generation of active/toxic metabolites, which may result in treatment discontinuation and inadequate patient compliance.
Topical formulations intended for dermal delivery are typically solutions, suspensions, gels, creams, ointments, lotions, sprays, and foams and can contain one or more conventional carriers as described herein. The formulation composition should be prepared with the goal of delivering the active ingredient to the appropriate layer(s) of the skin, minimizing systemic exposure, and preventing skin irritation. Additionally the pharmaceutical composition should be physically and chemically stable. Depending on the selected dosage form, one or more additional excipients as described herein may be necessary, e.g ., pH adjusting agents, chelating agents, preservatives, co-solvents, penetration enhancers, humectants, thickening, gelling, viscosity building agents, surfactants, propellants, fragrances, colorants, or any combination or mixture thereof.
In some embodiments, topical formulations can contain one or more conventional carriers as described herein. In some embodiments, ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white petrolatum, and the like. Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g. glycerinemonostearate, PEG- glycerinemonostearate and cetylstearyl alcohol. Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like. In some embodiments, topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention. The topical formulations can be suitably packaged in tubes of, for example, 100 g, which are optionally associated with instructions for the treatment of cutaneous lupus erythematosus (CLE).
The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from cutaneous lupus erythematosus (CLE) in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
The therapeutic dosage of a compound of the present application can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics ( e.g ., hydrophobicity), and the route of administration. For example, the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 pg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems. The compositions of the invention can further include one or more additional pharmaceutical agents, examples of which are listed hereinabove.
Kits
The present application also includes pharmaceutical kits useful, for example, in the treatment and/or prevention of cutaneous lupus erythematosus (CLE), which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a JAK1 inhibitor described herein. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
EXAMPLES
The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results.
All statistical analysis of in vitro experiments were performed with GraphPad prism software (version 7) using Kruskal -Wallis-Test and Mann-Whitney U test.
Gene expression was analyzed with Partek Flow genomic analysis software and Subio Platform software vl.22.5266 using Welch's /-test. Confidence intervals were determined at 95%. <0.05 was considered to be “significant” (*), ><0.01 to be “highly significant” (**). KEGG pathways were mapped to differentially expressed genes using DAVID v6.8 (Database for Annotation, Visualization and Integrated Discovery). Example 1. In vitro JAK Kinase Assay
JAK1 inhibitors that can be used for the treatment of cytokine-related diseases or disorders are tested for inhibitory activity of JAK targets according to the following in vitro assay described in Park el al. , Analytical Biochemistry 1999, 269 , 94-104.
The catalytic domains of human JAK1 (a.a. 837-1142), JAK2 (a. a. 828-1132) and JAK3 (a.a. 781-1124) with an N-terminal His tag are expressed using baculovirus in insect cells and purified. The catalytic activity of JAK1, JAK2 or JAK3 was assayed by measuring the phosphorylation of a biotinylated peptide. The phosphorylated peptide was detected by homogenous time resolved fluorescence (HTRF). ICsos of compounds are measured for each kinase in the 40 microL reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8) buffer with 100 mM NaCl, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA. For the 1 mM ICso measurements, ATP concentration in the reactions is 1 mM. Reactions are carried out at room temperature for 1 hour and then stopped with 20 pL 45 mM EDTA, 300 nM SA-APC, 6 nM Eu- Py20 in assay buffer (Perkin Elmer, Boston, MA). Binding to the Europium labeled antibody takes place for 40 minutes and HTRF signal was measured on a Fusion plate reader (Perkin Elmer, Boston, MA). The compounds in Table 1 were tested in this assay and shown to have the ICso values in Table 1.
Example 2. Activated JAK1 is Strongly Expressed in Human CLE Skin Lesions
To investigate the specific role of JAK 1 -mediated signaling in CLE, phosphorylated JAK1 (pJAKl) expression in lesional skin (SCLE and CDLE subsets) was compared to Lichen planus (LP) as well as healthy controls. All punch biopsies of the different inflammatory skin disorders (N=34) were taken for diagnostic purposes from active skin lesions. Healthy controls (N=9) were taken from unaffected skin taken from plastical surgery. Skin samples were fixed with 4% formalin overnight or fixed in frozen nitrogen and proceeded for immunohistochemistry or RNA isolation. RNA was processed by the Next Generation Sequencing (NGS) Core Facility of the Medical Faculty of the University of Bonn using the QuantSeq 3’- mRNA Library Prep Kit by Lexogen. Illumina HiSeq 2500 was used for RNA sequencing (Standard 3’RNA seq with 50 cycles) In CLE skin lesions, the expression of pJAKl was significantly increased in keratinocytes from stratum basale to stratum granulosum and in dermal infiltrating immune cells. It was also observed that pJAKl was significantly enhanced in Lichen planus, an autoimmune disease sharing common histological features with CLE.
Example 2. JAK/STAT-Associated Innate Inflammatory Pathways are Significantly Activated in Human CLE Skin
Samples of lesional skin from CLE patients were H&E stained to confirm the clinical diagnosis in each case by an experienced dermatopathologist. Immunohistochemistry was performed using the REAL™ Detection Systems with Fast Red as chromogen (Agilent, Santa Clara, USA) with specific antibodies for pJAK (ABIN196869, antibodies-online), CXCL10 (ab9807, Cambridge, UK), MxA (M143, Haller, Freiburg, Germany) and CD45 (550539, BD, New Jersey). The expression was scored semi quantitatively from 0 = weak to 3 = strong (Wenzel et al. J Pathol. 2005, 205:435-442). Immunofluorescence analyses of JAK1- phosphorylation detected by anti-rabbit Rhodamine Red-X (711-295-152; Jackson ImmunoResearch, Baltimore, MD, USA) and DAPI (D9542, Sigma- Aldrich) were performed using a high-resolution microscope (Axio Observer Zl, Zeiss, Germany).
Within CLE lesional skin expression analysis revealed a significant activation of genes associated with both innate and adaptive immune pathways compared to healthy controls. In particular, genes of LE-associated proinflammatory chemokines (CXCL10,9,11) and other IFN-regulated proteins (OASL, OAS2, Mxl) as well as key drivers in cell death and B-cell activation (CXCR3, CASP10, AIM2, TRAIL, BLyS) were highly expressed. JAK/STAT signaling is a critical regulator of inflammatory gene transcription, therefore gene expression of STAT1 in LE lesions was also significantly increased, as shown in FIG. 1C. Corresponding to individual genes upregulated innate immune pathways included JAK/STAT pathway and associated cytokine-/chemokine signaling as well as upstream TLR-dependent and -independent DAMP-recognition pathways, as shown in FIG. ID. Example 3. INCB039110 Significantly Inhibits JAKl-phosphorylation in Cultured Immortalized Human Keratinocytes
To analyze the functional principle and effect of JAK inhibition, the following established in vitro models of CLE were used. Immortalized keratinocytes (HaCaT), were acquired from CLS Cell Lines Service GmbH, Eppelheim, Germany), normal human epidermal keratinocytes (NHEKs, FC-0025) and Human epidermis equivalents (epiCS, CS-1001) from CellSystems, Troisdorf, Germany. These cell lines were cultured according the manufactures protocols. Cultured keratinocytes were stimulated with endogenous nucleic acids (eNA, 1,25 pg/mL) isolated from unstimulated keratinocytes using the “Genomic DNA from tissue“ kit (Machery- Nagel, Dueren, Germany). Lipofectamine 2000 (Invitrogen, Carlsbad, USA) functioned as a transfection reagent (2.5 pL/mL). INCB039110, as well as ruxolitinib (Selleckchem, Eching, Germany) were added at a final concentration of 1 pM; JAK3 selective FM-381 was used as recommended (100 nm) (see e.g ., Forster et al, Cell Chem. Biol. 2016, 23:1335-1340). All experiments were implemented in biological triplicates. Enzyme-linked immunosorbant assays for human CXCL10 (DY266-05 R&D systems) were performed using DuoSet Ancillary Reagent Kit 2 (DY008 R&D systems) according to the supplied protocol, measured by Synergy HT Multi- Detection Multiplate Reader (BioTek, Winooski, VT, USA) and read out with Gen5 software (version 1.11.5).
JAKl-phosphorylation was strongly enhanced within immortalized keratinocytes (HaCaT) after stimulation with eNA, corresponding with the findings in CLE skin lesions described above (see Examples 2-3). As shown in FIGs. 2A-2B, JAKl-specific INCB039110 and JAK 1/2-specific ruxolitinib significantly decreased the activation of JAKl within stimulated cells.
Example 4. Pharmacological JAKl Inhibition Blocks the Expression of CLE- typical Proinflammatory Cytokines and Pathway Molecules In Vitro
To investigate the efficacy of pharmacological JAKl inhibition, in vitro analyses in three different CLE-models were performed (i) NHEK-cells, (ii) HaCaT - cells and (iii) 3D epidermis equivalents. As shown in FIG. 2C, pharmacological JAKl inhibition induced a significant downregulation of genes encoding key drivers of innate inflammatory pathways such as IFN-regulated chemokines (CXCL10, CXCL11), cell death (TRAIL, AIM2, TREX1) and cross-talk to adaptive immune cells (BlyS) within primary keratinocytes (NHEK) compared to untreated eNA- inflamed cells. Associated downregulated KEGG pathways are listed in Table A. These results were confirmed in HaCaT-cells, where both JAK1- and JAK1/2 inhibitors decreased the protein expression of CXCL10 significantly.
Table A.
Figure imgf000055_0001
“KEGG pathways were classified using Database for Annotation, Visualization and Integrated Discovery (DAVID ver. 6.8). E-values were generated with EASE Score. Count: number of genes >2- fold downregulated in NHEK by INCB039110 within the respective KEGG pathway.
The in vitro data disclosed herein demonstrates that JAKl selective inhibitors are as potent as JAK1/2 inhibitors in suppression of CLE typical cytokines, as shown in FIG. 2D, and prohibit gene expression encoding proinflammatory chemokines (CXCLlO-11), lymphocyte activators (BLyS) (see e.g. , Wenzel et al, Exp. Dermatol. 2018, 27:95-97) and cell death promotors (TRAIL (see e.g., Zahn et al, Br. J. Dermatol. 2011, 165:1118-1123), AIM2, Caspase 10, TREX1). Interestingly, inhibition of JAK3 did not obtain a reduction of CXCL10 expression, as shown in FIG. 2D, which is a central mediator of CLE-typical “interface dermatitis”. Without being bound by theory, this could explain the earlier failure of the JAK3/SYK blocking agent R333 in a clinical CLE study (see e.g, Presto et al, Br. J. Dermatol. 2018, 178:1308-1314). In 3D epidermis equivalents, exposure of a JAK1 selective inhibitor to stimulated epiCS revealed a significantly reduced CXCL10 protein expression, consistent to findings described above, as shown in FIG. 2E.
Example 5. In Vivo Topical Application of INCB039110 Ameliorates CLE-like Lesions in Lupus-Prone TREXl Mice
TREX1 mice (generated on C57BL/6J background; Cancer Research Institute, London, UK) were bred and maintained under specific pathogen-free conditions at the animal core facility of UKB Bonn (HET, Bonn, Germany). TREX1 mice (n=8) were back-shaved and treated with 0,2% DNFB (l-Fluor-2,4- dinitrobenzol, Sigma Aldrich). 4 days later, UV-irridiation on 3 sequential days started with 450 mJ/cm2 UVB for 115 seconds per day using UV801KL (Waldmann, Villingen-Schwenningen, Germany). For 7 days 1% INCB039110 or vehicle solved in DMSO and olive oil (50 pL per mouse) were applied topically. Every day photos of mice were taken and every 2 days mice were weighed.
TREX1 _/ mice spontaneously developed CLE-like erythrosquamous and partly ulcerated skin lesions at a certain age which intensified after UVB -provocation. Topical treatment with JAKl-specific INCB039110 for seven days continuously improved lesional skin regarding erythema, induration, scaling and size leading to a significantly reduced lupus-skin-activity-score (adapted CLASI score) compared to placebo-treated mice, as shown in FIGs. 3A-3B. In addition, distinct histological features such as epidermal thickness and infiltrating dermal immune cells were significantly improved by JAK1 inhibition, as shown in FIG. 3C.
The data disclosed in Examples 2-5 demonstrates that JAKl-specific inhibition significantly decreases the expression of CLE-typical proinflammatory cytokines in vitro and in vivo. Topical application of a JAK1 selective inhibitor was highly effective in the treatment of CLE-like lesions in lupus prone mice, supporting their potential use in human CLE.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in the present disclosure, including all patent, patent applications, and publications, is incorporated herein by reference in its entirety.

Claims

WHAT IS CLAIMED IS:
1. A method of treating a disease selected from cutaneous lupus erythematosus (CLE) and Lichen planus (LP) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a JAK1 selective inhibitor selected from:
{l-{l-[3-Fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
4- { 3 -(Cyanomethyl)-3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 - yljazetidin- 1 -yl } -N-[4-fluoro-2-(trifluoromethyl)phenyl]piperidine- 1 -carboxamide;
[3 -[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- lH-pyrazol- l-yl]-l-(l-{[2- (trifluoromethyl)pyrimidin-4-yl]carbonyl}piperidin-4-yl)azetidin-3-yl]acetonitrile;
4-[3-(cyanomethyl)-3-(3',5'-dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l- yl]-2,5-difluoro-N-[(lS)-2,2,2-trifluoro-l-methylethyl]benzamide;
((2R,5S)-5-{2-[(lR)-l-hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin- 1 -yl }tetrahydro-2H-pyran-2-yl)acetonitrile;
3 -[ 1 -(6-chloropyridin-2-yl)pyrrolidin-3 -yl]-3 -[4-(7H-pyrrolo[2,3 -djpyrimidin- 4-yl)- lH-pyrazol- 1 -yljpropanenitrile;
3-(l-[l,3]oxazolo[5,4-b]pyridin-2-ylpyrrolidin-3-yl)-3-[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]propanenitrile;
4-[(4- { 3 -cyano-2-[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 - yl]propyl}piperazin-l-yl)carbonyl]-3-fluorobenzonitrile;
4-[(4- { 3 -cyano-2-[3 -(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrrol- 1 - yl]propyl}piperazin-l-yl)carbonyl]-3-fluorobenzonitrile;
[trans-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]-3-(4-{[2-
(trifluoromethyl)pyrimidin-4-yl]carbonyl}piperazin-l-yl)cyclobutyl]acetonitrile;
{trans-3-(4-{[4-[(3-hydroxyazetidin-l-yl)methyl]-6-(trifluoromethyl)pyridin- 2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- y 1 ] cy cl obuty 1 } acetonitril e;
{trans-3-(4-{[4-{[(2S)-2-(hydroxymethyl)pyrrolidin-l-yl]methyl}-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-
4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile; {trans-3-(4-{[4-{[(2R)-2-(hydroxymethyl)pyrrolidin-l-yl]methyl}-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-
4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile;
4-(4- { 3 - [(dimethyl amino)methyl] -5 -fluorophenoxy } piped din- 1 -yl)-3-[4-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]butanenitrile;
5-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl } -N-isopropylpyrazine-2-carboxamide;
4-{3-(cyanomethyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl } -2,5-difluoro-N-[( 1 S)-2,2,2-trifluoro- 1 -methylethyljbenzamide;
5-{3-(cyanomethyl)-3-[4-(lH-pyrrolo[2,3-b]pyridin-4-yl)-lH-pyrazol-l- yljazetidin- 1 -yl } -N-isopropylpyrazine-2-carboxamide;
{l-(cis-4-{[6-(2-hydroxyethyl)-2-(trifluoromethyl)pyrimidin-4- yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin- 3 -yl} acetonitrile;
{l-(cis-4-{[4-[(ethylamino)methyl]-6-(trifluoromethyl)pyridin-2- yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin- 3 -yl} acetonitrile;
(l-(cis-4-{[4-(l-hydroxy-l -methyl ethyl)-6-(trifluoromethyl)pyri din-2- yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-
3 -yl} acetonitrile;
(l-(cis-4-{[4-{[(3R)-3-hydroxypyrrolidin-l-yl]methyl}-6-
(trifluoromethyl)pyridin-2-yl]oxy}cyclohexyl)-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{ 1 -(ci s-4- { [4- { [(3 S)-3 -hy droxypyrrolidin- 1 -yl] methyl }-6- (trifluoromethyl)pyridin-2-yl]oxy}cyclohexyl)-3-[4-(7H-pynOlo[2,3-d]pyrimidin-4- yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile;
{trans-3-(4-{[4-({[(lS)-2-hydroxy-l-methylethyl]amino}methyl)-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-
4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile;
(trans-3-(4-{[4-({[(2R)-2-hydroxypropyl]amino}methyl)-6-
(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l-[4-(7H-pyrrolo[2,3-d]pyrimidin-
4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile{trans-3-(4-{[4-({[(2S)-2- hydroxypropyl]amino}methyl)-6-(trifluoromethyl)pyridin-2-yl]oxy}piperidin-l-yl)-l- [4-(7H-pyirolo[2,3-d]pyrimidin-4-yl)-lH-pyrazol-l-yl]cyclobutyl}acetonitrile; and {trans-3 -(4- { [4-(2 -hydroxy ethyl)-6-(trifluoromethyl)pyridin-2- yl]oxy }piperidin- 1 -yl)- 1 -[4-(7H-pyrrolo[2,3 -d]pyrimidin-4-yl)- lH-pyrazol- 1 - y 1 ] cy cl obuty 1 } acetonitril e; or a pharmaceutically acceptable salt of any of the aforementioned.
2. The method of claim 1, wherein the JAK1 selective inhibitor, or a pharmaceutically acceptable salt thereof, is selective for JAK1 over JAK2, JAK3, and TYK2.
3. The method of claim 1 or 2, wherein the JAK1 selective inhibitor is { l-{ l-[3- fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
4. The method of claim 3, wherein the pharmaceutically acceptable salt is { l-{ 1- [3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3[4-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-lH-pyrazol-l-yl]azetidin-3-yl}acetonitrile adipic acid salt.
5. The method of claim 1 or 2, wherein the JAK1 selective inhibitor is 4-[3- (cyanomethyl)-3-(3',5'-dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5- difluoro-N-[(lS)-2,2,2-trifluoro-l-methylethyl]benzamide, or a pharmaceutically acceptable salt thereof.
6. The method of claim 5, wherein the pharmaceutically acceptable salt is 4-[3- (cyanomethyl)-3-(3',5'-dimethyl-lH,rH-4,4'-bipyrazol-l-yl)azetidin-l-yl]-2,5- difluoro-N-[(lS)-2,2,2-trifluoro-l-methylethyl]benzamide phosphoric acid salt.
7. The method of claim 1 or 2, wherein the JAK1 selective inhibitor is ((2R,5S)- 5-{2-[(lR)-l-hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl}tetrahydro- 2H-pyran-2-yl)acetonitrile, or a pharmaceutically acceptable salt thereof.
8. The method of claim 1 or 2, wherein the JAK1 selective inhibitor is ((2R,5S)- 5-{2-[(lR)-l-hydroxyethyl]-lH-imidazo[4,5-d]thieno[3,2-b]pyridin-l-yl}tetrahydro- 2H-pyran-2-yl)acetonitrile monohydrate.
9. The method of any one of claims 1 to 8, further comprising administering to the patient an additional therapeutic agent.
10. The method of claim 9, wherein the additional therapeutic agent is selected from a JAK1/JAK2 inhibitor, a JAK1/JAK3 inhibitor, a TYK2 inhibitor, or a pharmaceutically acceptable salt of any of the aforementioned.
11. The method of claim 10, wherein the additional therapeutic agent is a JAK1/JAK2 inhibitor, or a pharmaceutically acceptable salt thereof.
12. The method of claim 10 or 11, wherein the JAK1/JAK2 inhibitor, or a pharmaceutically acceptable salt thereof, is selective for JAK1 and JAK2 over JAK3 and TYK2.
13. The method of any one of claims 10 to 12, wherein the JAK1/JAK2 inhibitor is ruxolitinib, or a pharmaceutically acceptable salt thereof.
14. The method of claim 13, wherein the method comprises topical administration of the ruxolitinib, or a pharmaceutically acceptable salt thereof, to the patient.
15. The method of claim 13, wherein the method comprises oral administration of the ruxolitinib, or a pharmaceutically acceptable salt thereof, to the patient.
16. The method of any one of claims 13 or 15, wherein the pharmaceutically acceptable salt is ruxolitinib phosphate.
17. The method of any one of claims 10 to 12, wherein the JAK1/JAK2 inhibitor is ruxolitinib, or a pharmaceutically acceptable salt thereof, wherein one or more hydrogen atoms are replaced by deuterium atoms.
18. The method of claim 10, wherein the additional therapeutic agent is a JAK1/JAK3 inhibitor, or a pharmaceutically acceptable salt thereof.
19. The method of claim 10 or 18, wherein the JAK1/JAK3 inhibitor is tofacitinib, or a pharmaceutically acceptable salt thereof.
20. The method of any one of claims 1 to 19, wherein the method comprises topical administration of the JAK1 selective inhibitor to the patient.
21. The method of claim any one of claims 1 to 19, wherein the method comprises oral administration of the JAK1 selective inhibitor to the patient.
22. The method of any one of claims 1 to 21, wherein the disease is cutaneous lupus erythematosus (CLE).
23. The method of claim 22, wherein the cutaneous lupus erythematosus (CLE) is selected from subacute cutaneous lupus erythematosus (SCLE) and chronic discoid lupus erythematosus (CDLE).
24. The method of any one of claims 1 to 21, wherein the disease is Lichen planus (LP).
PCT/US2019/056533 2019-10-16 2019-10-16 Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp) WO2021076124A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2022523053A JP2023506118A (en) 2019-10-16 2019-10-16 Use of JAK1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (LP)
PCT/US2019/056533 WO2021076124A1 (en) 2019-10-16 2019-10-16 Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2019/056533 WO2021076124A1 (en) 2019-10-16 2019-10-16 Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp)

Publications (1)

Publication Number Publication Date
WO2021076124A1 true WO2021076124A1 (en) 2021-04-22

Family

ID=68470631

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/056533 WO2021076124A1 (en) 2019-10-16 2019-10-16 Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp)

Country Status (2)

Country Link
JP (1) JP2023506118A (en)
WO (1) WO2021076124A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022072814A1 (en) * 2020-10-02 2022-04-07 Incyte Corporation Topical ruxolitinib for treating lichen planus
WO2022235613A1 (en) * 2021-05-03 2022-11-10 Incyte Corporation Jak1 pathway inhibitors for the treatment of prurigo nodularis

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO2000009495A1 (en) 1998-08-11 2000-02-24 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
WO2000053595A1 (en) 1999-03-06 2000-09-14 Astrazeneca Ab Pyrimidine compounds
WO2001014402A1 (en) 1999-08-19 2001-03-01 Isis Pharmaceuticals, Inc. Antisense modulation of focal adhesion kinase expression
WO2001064655A1 (en) 2000-03-01 2001-09-07 Astrazeneca Ab 2, 4-di(hetero-)arylamino (-oxy)-5-substituted pyrimidines as antineoplastic agents
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2003024967A2 (en) 2001-09-19 2003-03-27 Aventis Pharma S.A. Indolizines as kinase protein inhibitors
WO2003037347A1 (en) 2001-10-30 2003-05-08 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
WO2003099771A2 (en) 2002-05-29 2003-12-04 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2004046120A2 (en) 2002-11-15 2004-06-03 Vertex Pharmaceuticals Incorporated Diaminotriazoles useful as inhibitors of protein kinases
WO2004056786A2 (en) 2002-12-20 2004-07-08 Pfizer Products Inc. Pyrimidine derivates for the treatment of abnormal cell growth
WO2004080980A1 (en) 2003-03-14 2004-09-23 Novartis Ag 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2005028444A1 (en) 2003-09-24 2005-03-31 Novartis Ag 1,4-disubstituted isoquinilone derivatives as raf-kinase inhibitors useful for the treatment of proliferative diseases
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
US7598257B2 (en) 2005-12-13 2009-10-06 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US20100298334A1 (en) 2009-05-22 2010-11-25 Rodgers James D N-(HETERO)ARYL-PYRROLIDINE DERIVATIVES OF PYRAZOL-4-YL-PYRROLO[2,3-d]PYRIMIDINES AND PYRROL-3-YL-PYRROLO[2,3-d]PYRIMIDINES AS JANUS KINASE INHIBITORS
US20110059951A1 (en) 2009-09-01 2011-03-10 Rodgers James D HETEROCYCLIC DERIVATIVES OF PYRAZOL-4-YL-PYRROLO[2,3-d]PYRIMIDINES AS JANUS KINASE INHIBITORS
US20110224190A1 (en) 2010-03-10 2011-09-15 Taisheng Huang Piperidin-4-yl azetidine derivatives as jak1 inhibitors
US20120149682A1 (en) 2010-11-19 2012-06-14 Rodgers James D Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
US20120149681A1 (en) 2010-11-19 2012-06-14 Rodgers James D Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US20130018034A1 (en) 2011-06-20 2013-01-17 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
US20130045963A1 (en) 2011-08-18 2013-02-21 Incyte Corporation Cyclohexyl Azetidine Derivatives as JAK Inhibitors
US20130060026A1 (en) 2011-09-07 2013-03-07 Incyte Corporation Processes and intermediates for making a jak inhibitor
US20140005166A1 (en) 2012-05-18 2014-01-02 Incyte Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US20140121198A1 (en) 2012-11-01 2014-05-01 Incyte Corporation Tricyclic fused thiophene derivatives as jak inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US20140256941A1 (en) 2013-03-06 2014-09-11 Incyte Corporation Processes and intermediates for making a jak inhibitor
US20140343030A1 (en) 2013-05-17 2014-11-20 Incyte Corporation Bipyrazole derivatives as jak inhibitors
US20150344497A1 (en) 2014-04-30 2015-12-03 Incyte Corporation Processes of preparing a jak1 inhibitor and new forms thereto
US9249149B2 (en) 2012-06-15 2016-02-02 Concert Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib
US9540367B2 (en) 2012-08-17 2017-01-10 Concert Pharmaceuticals, Inc. Deuterated baricitinib
WO2018087202A1 (en) * 2016-11-10 2018-05-17 Galapagos Nv Compounds and pharmaceutical compositions thereof for the treatment of inflammatory diseases
WO2019191679A1 (en) * 2018-03-30 2019-10-03 Incyte Corporation Biomarkers for inflammatory skin disease

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7096592B2 (en) * 2016-02-16 2022-07-06 ワシントン・ユニバーシティ JAK inhibitors and their use

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO2000009495A1 (en) 1998-08-11 2000-02-24 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
WO2000053595A1 (en) 1999-03-06 2000-09-14 Astrazeneca Ab Pyrimidine compounds
WO2001014402A1 (en) 1999-08-19 2001-03-01 Isis Pharmaceuticals, Inc. Antisense modulation of focal adhesion kinase expression
WO2001064655A1 (en) 2000-03-01 2001-09-07 Astrazeneca Ab 2, 4-di(hetero-)arylamino (-oxy)-5-substituted pyrimidines as antineoplastic agents
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2003024967A2 (en) 2001-09-19 2003-03-27 Aventis Pharma S.A. Indolizines as kinase protein inhibitors
WO2003037347A1 (en) 2001-10-30 2003-05-08 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
WO2003099771A2 (en) 2002-05-29 2003-12-04 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2004046120A2 (en) 2002-11-15 2004-06-03 Vertex Pharmaceuticals Incorporated Diaminotriazoles useful as inhibitors of protein kinases
WO2004056786A2 (en) 2002-12-20 2004-07-08 Pfizer Products Inc. Pyrimidine derivates for the treatment of abnormal cell growth
WO2004080980A1 (en) 2003-03-14 2004-09-23 Novartis Ag 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2005028444A1 (en) 2003-09-24 2005-03-31 Novartis Ag 1,4-disubstituted isoquinilone derivatives as raf-kinase inhibitors useful for the treatment of proliferative diseases
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
US7598257B2 (en) 2005-12-13 2009-10-06 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US20100298334A1 (en) 2009-05-22 2010-11-25 Rodgers James D N-(HETERO)ARYL-PYRROLIDINE DERIVATIVES OF PYRAZOL-4-YL-PYRROLO[2,3-d]PYRIMIDINES AND PYRROL-3-YL-PYRROLO[2,3-d]PYRIMIDINES AS JANUS KINASE INHIBITORS
US20110059951A1 (en) 2009-09-01 2011-03-10 Rodgers James D HETEROCYCLIC DERIVATIVES OF PYRAZOL-4-YL-PYRROLO[2,3-d]PYRIMIDINES AS JANUS KINASE INHIBITORS
US20110224190A1 (en) 2010-03-10 2011-09-15 Taisheng Huang Piperidin-4-yl azetidine derivatives as jak1 inhibitors
US20120149682A1 (en) 2010-11-19 2012-06-14 Rodgers James D Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
US20120149681A1 (en) 2010-11-19 2012-06-14 Rodgers James D Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US20130018034A1 (en) 2011-06-20 2013-01-17 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
US20130045963A1 (en) 2011-08-18 2013-02-21 Incyte Corporation Cyclohexyl Azetidine Derivatives as JAK Inhibitors
US20130060026A1 (en) 2011-09-07 2013-03-07 Incyte Corporation Processes and intermediates for making a jak inhibitor
US20140005166A1 (en) 2012-05-18 2014-01-02 Incyte Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US9249149B2 (en) 2012-06-15 2016-02-02 Concert Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib
US9540367B2 (en) 2012-08-17 2017-01-10 Concert Pharmaceuticals, Inc. Deuterated baricitinib
US20140121198A1 (en) 2012-11-01 2014-05-01 Incyte Corporation Tricyclic fused thiophene derivatives as jak inhibitors
US20140256941A1 (en) 2013-03-06 2014-09-11 Incyte Corporation Processes and intermediates for making a jak inhibitor
US20140343030A1 (en) 2013-05-17 2014-11-20 Incyte Corporation Bipyrazole derivatives as jak inhibitors
US20150344497A1 (en) 2014-04-30 2015-12-03 Incyte Corporation Processes of preparing a jak1 inhibitor and new forms thereto
WO2018087202A1 (en) * 2016-11-10 2018-05-17 Galapagos Nv Compounds and pharmaceutical compositions thereof for the treatment of inflammatory diseases
WO2019191679A1 (en) * 2018-03-30 2019-10-03 Incyte Corporation Biomarkers for inflammatory skin disease

Non-Patent Citations (85)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
A KUHN ET AL: "Advances in the treatment of cutaneous lupus erythematosus", LUPUS, vol. 25, no. 8, 31 May 2016 (2016-05-31), GB, pages 830 - 837, XP055462245, ISSN: 0961-2033, DOI: 10.1177/0961203316641771 *
A. KEREKES, J. MED. CHEM., vol. 54, 2011, pages 201 - 210
ALAN F. THOMAS: "Deuterium Labeling in Organic Chemistry", 1971, APPLETON-CENTURY-CROFTS
ALBRECHT ET AL., J. INVEST. DERMATOL., vol. 125, no. 5, November 2005 (2005-11-01), pages 889 - 94
ALBRECHT J ET AL., J. INVEST. DERMATOL., vol. 125, 2005, pages 889 - 94
ALVES DE MEDEIROS ET AL., PLOS ONE, vol. 11, 2016, pages e0164080
ARRUE ET AL., J. CUTAN. PATHOL., vol. 34, no. 1, 2007, pages 18 - 21
BANEIJEE ET AL., DRUGS, vol. 77, 2017, pages 521 - 546
BRIAND ET AL., ANN. RHEUM. DIS., vol. 78, 2019, pages 431 - 433
CAPRONI ET AL., INT. J. DERMATOL., vol. 40, no. 1, 2001, pages 59 - 62
CARICCHIO ET AL., THE JOURNAL OF IMMUNOLOGY, vol. 171, 2003, pages 5778 - 5786
CHEN ET AL., FLOOORES., 2019, pages 8
CHONG ET AL., BR. J. DERMATOL., vol. 166, no. 1, 2012, pages 29 - 35
CROWSONMAGRO, J. CUTAN. PATHOL., vol. 28, no. 1, 2001, pages 1 - 23
FABBRI ET AL., AM. J. CLIN. DERMATOL., vol. 4, no. 7, 2003, pages 449 - 65
FONESCA ET AL., AUTOIMMUNITY REVIEWS, vol. 8, 2009, pages 538 - 42
FORSTER ET AL., CELL CHEM. BIOL., vol. 23, 2016, pages 1335 - 1340
FURIE ET AL., ARTHRITIS & RHEUMATOLOGY, vol. 69, 2017, pages 376 - 386
FURIE ET AL., ARTHRITIS RHEUMATOL., vol. 69, 2017, pages 376 - 86
GRIEVES ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 112, 2015, pages 5117 - 5122
GUNTHER ET AL., DERMATOLOGY (BASEL), vol. 219, 2009, pages 162 - 166
GUNTHER ET AL., J. AM. ACAD. DERMATOL., vol. 69, 2013, pages el79 - 81
GUSCHIN ET AL., EMBO J, vol. 14, 1995, pages 1421
HEDRICH ET AL., CLIN. RHEUMATOL., vol. 27, no. 8, 2008, pages 949 - 54
HORNUNG ET AL., N. ENGL. J. MED., vol. 371, 2014, pages 2537 - 2538
HORNUNG ET AL., N. ENGL. J. MED., vol. 372, 2015, pages 1274
HOWELL ET AL., ANN. ALLERGY ASTHMA IMMUNOL., vol. 120, 2018, pages 367 - 375
JENS ATZRODTVOLKER DERDAUTHORSTEN FEYJOCHEN ZIMMERMANN: "The Renaissance of H/D Exchange", ANGEW. CHEM. INT. ED., 2007, pages 7744 - 7765, XP055192405, DOI: 10.1002/anie.200700039
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2
KALUNIAN ET AL., ANN. RHEUM. DIS., vol. 75, 2016, pages 196 - 202
KATAYAMA ET AL., J. INVEST. DERMATOL., vol. 138, 2018, pages 1785 - 1794, Retrieved from the Internet <URL:https://doi.org/10.1016/jjid.2018.08.017>
KHAMASHTA ET AL., ANN. RHEUMAT. DIS., vol. 75, 2016, pages 1909 - 16
KLAESCHEN ET AL., EXP. DERMATOL., vol. 26, 2017, pages 728 - 730
KLEIN ET AL., ARCH. DERMATOL., vol. 147, 2011, pages 203 - 8
KONIG ET AL., ANN. RHEUM. DIS., vol. 76, no. 2, 2017, pages 468 - 472
KUHN ET AL., ARTHRITIS RHEUM., vol. 54, 2006, pages 939 - 950
KUHN ET AL., SEMIN. IMMUNOPATHOL., vol. 38, 2016, pages 97 - 112
KUHNLANDMANN, J. AUTOIMMUN., vol. 48-49, 2014, pages 14 - 19
KUNZ ET AL., EXP. DERMATOL., vol. 24, 2015, pages 510 - 515
MAHAJAN ET AL., FRONT IMMUNOL., vol. 7, 2016
MELLER ET AL., ARTHRITIS RHEUM., vol. 52, 2005, pages 1504 - 1516
MERRILL ET AL., ANN. RHEUM. DIS., vol. 70, 2011, pages 1905 - 1913
MIKITA ET AL., J. DERMATOL., vol. 38, 2011, pages 839 - 849
MORGAN ET AL., ANNU. REV. MED., vol. 59, 2008, pages 213 - 222
MUSTELIN ET AL., FRONT. IMMUNOL., vol. 10, 2019, pages 238
OGUNSANYA ET AL., INT. J. WOMENS DERMATOL., vol. 4, 2018, pages 152 - 158
PARDANANI ET AL., LEUKEMIA, vol. 22, 2008, pages 23 - 30
PARK ET AL., ANALYTICAL BIOCHEMISTRY, vol. 269, 1999, pages 94 - 104
PATRIA INACIO: "Cutaneous Lupus Erythematosus Patients May Benefit from JAK1 Inhibitors", INTERNET CITATION, 17 October 2016 (2016-10-17), pages 1, XP002779504, Retrieved from the Internet <URL:https://lupusnewstoday.com/2016/10/17/jak1-inhibitors-may-prove-beneficial-cutaneous-lupus-erythematosus/> [retrieved on 20180323] *
PESCHKE ET AL., J. INVEST. DERMATOL., vol. 134, 2014, pages 1456 - 1459
PRESTO ET AL., BR. J. DERMATOL., vol. 178, 2018, pages 1308 - 1314
R. XU, J. LABEL COMPD. RADIOPHARM., vol. 58, 2015, pages 308 - 312
RONNBLOM ET AL., ACTA ONCOL., vol. 30, 1991, pages 537 - 540
ROTHFIELD ET AL., CLIN. DERMATOL., vol. 24, no. 5, September 2006 (2006-09-01), pages 348 - 62
SAADEH ET AL., EXP. DERMATOL., vol. 25, 2016, pages 415 - 421
SAMOTIJ ET AL., POSTEPY DERMATOL. ALERGOL., vol. 35, 2018, pages 192 - 198
SANTOSVERSTOVSEK, EXPERT OPIN. PHARMACOTHER., vol. 15, 2014, pages 1465 - 1473
SARKAR ET AL., ANN. RHEUM. DIS., vol. 77, 2018, pages 1653 - 1664
SCHOLTISSEK ET AL., J. INVEST. DERMATOL., vol. 137, 2017, pages 1484 - 1492
SCHWARTZ ET AL., NAT. REV. DRUG DISCOV., vol. 16, 2017, pages 843 - 862
SCHWARTZ ET AL., NAT. REV. RHEUMATOL., vol. 12, 2016, pages 25 - 36
SHREBERK-HASSIDIM ET AL., J. AM. ACAD. DERMATOL., vol. 76, 2017, pages 745 - 753
SINHADEY-RAO, J. INVESTIG. DERMATOL. SYMP. PROC., vol. 18, 2017, pages S75 - S80
SMOLEN ET AL., LANCET, vol. 371, 2008, pages 987
SPOERL ET AL., BLOOD, vol. 123, 2014, pages 3832 - 3842
SRIVASTAVA ET AL., ACTA. DERM. VENEREOL., vol. 98, 2018, pages 772 - 775
SZCZ CH ET AL., LUPUS, vol. 26, 2017, pages 791 - 807
TEBBEORFANOS, LUPUS, vol. 6, no. 2, 1997, pages 96 - 104
VAN VOLLENHOVEN ET AL., THE LANCET, vol. 392, 2018, pages 1330 - 9
WALLACE ET AL., LANCET, vol. 392, no. 10143, 2018, pages 222 - 231
WALLINGSONTHEIMER, AM. J. CLIN. DERMATOL., vol. 10, no. 6, 2009, pages 365 - 81
WELSCH ET AL., EUR. J. IMMUNOL., vol. 47, 2017, pages 1096 - 1107
WENZEL ET AL., ARCH. DERMATOL. RES., vol. 301, 2009, pages 83 - 86
WENZEL ET AL., BR. J. DERMATOL., vol. 157, 2007, pages 752 - 757
WENZEL ET AL., EXP. DERMATOL., vol. 27, 2018, pages 95 - 97
WENZEL ET AL., J. AM. ACAD. DERMATOL., vol. 58, 2008, pages 437 - 442
WENZEL ET AL., J. INVEST. DERMATOL., vol. 136, 2016, pages 1281 - 1283
WENZEL ET AL., J. PATHOL., vol. 205, 2005, pages 435 - 442
WENZELTUTING, J. INVEST. DERMATOL., vol. 128, 2008, pages 2392 - 2402
WERTHMERRILL, BR. J. DERMATOL., vol. 180, no. 5, 2019, pages 964 - 965
WOLLENBERG ET AL., J. INVEST. DERMATOL., vol. 119, 2002, pages 1096 - 1102
YU ET AL., J. AUTOIMMUN., vol. 41, 2013, pages 34 - 45
ZAHN ET AL., BR. J. DERMATOL., vol. 165, 2011, pages 1118 - 1123
ZIMMERMAN ET AL., JAMA DERMATOL.

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022072814A1 (en) * 2020-10-02 2022-04-07 Incyte Corporation Topical ruxolitinib for treating lichen planus
WO2022235613A1 (en) * 2021-05-03 2022-11-10 Incyte Corporation Jak1 pathway inhibitors for the treatment of prurigo nodularis

Also Published As

Publication number Publication date
JP2023506118A (en) 2023-02-15

Similar Documents

Publication Publication Date Title
US9498467B2 (en) Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
ES2829914T3 (en) Treatment of malignant B cell diseases using a combination of JAK and PI3K inhibitor
US20220241286A1 (en) Treatment of hidradenitis suppurativa using jak inhibitors
US11833152B2 (en) JAK1 pathway inhibitors for the treatment of cytokine-related disorders
CA2921568A1 (en) Survival benefit in patients with solid tumors with elevated c-reactive protein levels
EP2527344A1 (en) Pyridin-2(1H)-one derivatives useful as medicaments for the treatment of myeloproliferative disorders, transplant rejection, immune-mediated and inflammatory diseases
EP2463289A1 (en) Imidazo[1,2-b]pyridazine derivatives as JAK inhibitors
CN114007621A (en) JAK1 pathway inhibitors for the treatment of chronic lung allograft dysfunction
WO2021076124A1 (en) Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp)
KR20240009964A (en) JAK1 pathway inhibitors for the treatment of prurigo nodosum
WO2015091531A1 (en) Imidazolopyrimidin-2-yl derivatives as jak inhibitors
US20210113566A1 (en) Use of jak1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (lp)
KR20230118118A (en) JAK1 pathway inhibitors for the treatment of vitiligo
US20240058343A1 (en) Treatment of urticaria using jak inhibitors
TWI835786B (en) Treatment of hidradenitis suppurativa using jak inhibitors
KR20230157307A (en) Combination therapy involving JAK pathway inhibitors and ROCK inhibitors
EA042956B1 (en) JAK1 PATHWAY INHIBITORS FOR THE TREATMENT OF CYTOKINE RELATED DISORDERS
EP2360158A1 (en) Pyrazole derivatives as jak inhibitors

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: 19798789

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022523053

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19798789

Country of ref document: EP

Kind code of ref document: A1