Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INDOLINE DERIVATIVES, COMPOSITIONS COMPRISING THEM AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2017/125932
Kind Code:
A1
Abstract:
The present invention is directed to indoline derivatives and salts thereof, compositions comprising them and uses thereof for the treatment of diseases and disorders associated with at least one of oxidative stress, an immune response, release of NO and release of pro-inflammatory cytokine.

Inventors:
WEINSTOCK-ROSIN MARTA (IL)
NUDELMAN ABRAHAM (IL)
ZEELI SHANI (IL)
Application Number:
PCT/IL2017/050079
Publication Date:
July 27, 2017
Filing Date:
January 19, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
YISSUM RES DEV CO (IL)
UNIV BAR ILAN (IL)
International Classes:
C07D209/08; A61K31/403; A61P25/00; C07D209/14; C07D209/18
Domestic Patent References:
WO2013150529A22013-10-10
WO2015049667A12015-04-09
WO2001040183A12001-06-07
Foreign References:
US6002018A1999-12-14
US6380238B12002-04-30
US20110112148A12011-05-12
US5206240A1993-04-27
RU2387640C12010-04-27
Other References:
R. R. HUNT ET AL: "A new synthesis of methoxyindoles", JOURNAL OF THE CHEMICAL SOCIETY, SECTION C: ORGANIC CHEMISTRY.>6015C, 1966, GB, pages 344, XP055349302, ISSN: 0022-4952, DOI: 10.1039/j39660000344
PAPAGEORGIOU G ET AL: "Effects of Aromatic Substituents on the Photocleavage of 1-Acyl-7-nitroindolines", TETRAHEDRON, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 56, no. 41, 6 October 2000 (2000-10-06), pages 8197 - 8205, XP004217694, ISSN: 0040-4020, DOI: 10.1016/S0040-4020(00)00745-6
LIANGZHEN CAI ET AL: "Palladium-catalyzed Coupling between Aryl Halides and Trimethylsilylacetylene Assisted by Dimethylaminotrimethyltin", CHINESE JOURNAL OF CHEMISTRY, vol. 29, no. 5, 2011, CN, pages 1059 - 1062, XP055349512, ISSN: 1001-604X, DOI: 10.1002/cjoc.201190180
JIHYE PARK ET AL: "Mild Rh(III)-Catalyzed C7-Allylation of Indolines with Allylic Carbonates", THE JOURNAL OF ORGANIC CHEMISTRY, vol. 80, no. 3, 6 February 2015 (2015-02-06), pages 1818 - 1827, XP055349518, ISSN: 0022-3263, DOI: 10.1021/jo502733q
HENRY RAPOPORT ET AL: "Alkaloids of Geissospermum vellosii. Further Studies on Geissospermine and the Structures of the Indolic Cleavage Products, Geissoschizine 1 and Apogeissoschizine", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 82, no. 16, 1 August 1960 (1960-08-01), pages 4404 - 4414, XP055354489, ISSN: 0002-7863, DOI: 10.1021/ja01501a069
RICHARD A. GLENNON ET AL: "Synthesis and evaluation of a novel series of N,N-dimethylisotryptamines", JOURNAL OF MEDICINAL CHEMISTRY, vol. 27, no. 1, 1 January 1984 (1984-01-01), US, pages 41 - 45, XP055354516, ISSN: 0022-2623, DOI: 10.1021/jm00367a008
ADLER S. ET AL.: "Induction of severe systemic lupus erythematosus by TNF blockade and response to anti-IL-6 strategy", J ALLERGY CLIN IMMUNOL, vol. 131, 2013, pages 1235 - 1237
ALLEZ M. ET AL.: "Report of the ECCO pathogenesis workshop on anti-TNF therapy failures in inflammatory bowel diseases: definitions, frequency and pharmacological aspects", J CROHNS COLITIS, vol. 4, no. 4, 2010, pages 355 - 366, XP027375502
AMUNDSEN LH; NELSON LS: "Reduction of nitriles to primary amines with lithium aluminum hydride", J AM CHEM SOC, vol. 73, 1951, pages 242 - 244
ASTIL B; BOEKELHEIDE V: "Synthesis of 5-Ketolilolidine", J ORG CHEM, vol. 23, 1958, pages 316 - 318
BAUMANN H; GAULDIE J: "The acute phase response", IMMUNOL TODAY, vol. 15, 1994, pages 74 - 80, XP023937953, DOI: doi:10.1016/0167-5699(94)90137-6
BEN-HORIN S; CHOWERS Y: "Tailoring anti-TNF therapy in IBD: drug levels and disease activity", NAT REV GASTROENTEROL HEPATOL, vol. LL, no. 4, 2014, pages 243 - 255
BERNEY T. ET AL: "Serum profiles of interleukin-6, interleukin-8, and interleukin-10 in patients with severe and mild acute pancreatitis", PANCREAS, vol. 18, 1999, pages 371 - 377
BONGARTZ T. ET AL: "Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials", JAMA, vol. 295, 2006, pages 2275 - 2285, XP002720114, DOI: doi:10.1001/jama.295.19.2275
CORNU M. ET AL: "mTOR in aging, metabolism, and cancer", CURR OP GEN DEV, vol. 23, 2013, pages 53 - 62
DEFURIA J. ET AL.: "B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile", PROC NATL ACAD SCI USA, vol. 110, 2013, pages 5133 - 5138
DELLAGIOIA N. ET AL: "Bupropion pretreatment of endotoxin-induced depressive symptoms", BRAIN BEHAV IMMUN, vol. 31, 2013, pages 197 - 204
DHIR A. ET AL: "Involvement of L-arginine-nitric oxide-cyclic guanosine monophosphate pathway in the antidepressant-like effect of venlafaxine in mice", PROG NEUROPSYCHOPHARMACOL BIOL PSYCHIATRY, vol. 31, 2007, pages 921 - 925, XP022054059, DOI: doi:10.1016/j.pnpbp.2007.02.008
DIAS V. ET AL: "The role of oxidative stress in Parkinson's disease", J PARKINSON'S DIS, vol. 3, 2013, pages 461 - 491
DINARELLO CA: "Pro-inflammatory and anti-inflammatory cytokines as mediators in the pathogenesis of septic shock", CHEST, vol. 112, 1997, pages 321S - 329S
GONZALEZ-JUANATEY C. ET AL.: "Anti-TNF-alpha-adalimumab therapy is associated with persistent improvement of endothelial function without progression of carotid intima-media wall thickness in patients with rheumatoid arthritis refractory to conventional therapy", MEDIATORS INFLAMM, 2012, pages 674265
HAJJAR D.P. ET AL: "Biological relevance of inflammation and oxidative stress in the pathogenesis of arterial diseases", AM J PATHOL, vol. 182, 2013, pages 1474 - 1481
IRONY-TUR-SINAI M. ET AL: "Amelioration of autoimmune neuroinflammation by recombinant human alpha-fetoprotein", EXP NEUROL, vol. 198, 2006, pages 136 - 144, XP024945737, DOI: doi:10.1016/j.expneurol.2005.11.012
KANASAKI K. ET AL: "Diabetic nephropathy: the role of inflammation in fibroblast activation and kidney fibrosis", FRONT ENDOCRINOL (LAUSANNE)., vol. 4, 6 February 2013 (2013-02-06), pages 7 - 15
LEVITT J.E. ET AL: "Treatment of acute lung injury: historical perspective and potential future therapies", SEMIN RESPIR CRIT CARE MED, vol. 27, 2006, pages 426 - 437
MAES M.: "Major depression and activation of the inflammatory response system", ADV EXP MED BIOL, vol. 461, 1999, pages 25 - 46
MAES M.: "Inflammatory and oxidative and nitrosative stress cascades as new drug targets in myalgic encephalomyelitis and chronic fatigue syndrome", MOD TRENDS PHARMACOPSYCHIATRY, vol. 28, 2013, pages 162 - 174
MAGRO F; PORTELA F: "Management of inflammatory bowel disease with infliximab and other anti-tumor necrosis factor alpha therapies", BIODRUGS, vol. 24, no. 1, 2010, pages 3 - 14, XP009186432, DOI: doi:10.2165/11586290-000000000-00000
MANDELL BF; SOBELL JM: "The role of TNF inhibitors in psoriatic disease", SEMIN CUTAN MED SURG, vol. 33, 2014, pages 64 - 68
MANGIALASCHE F. ET AL.: "Biomarkers of oxidative and nitrosative damage in Alzheimer's disease and mild cognitive impairment", AGEING RES REV, vol. 8, 2009, pages 285 - 305, XP026520063, DOI: doi:10.1016/j.arr.2009.04.002
MARTINEZ-CASTELAO A. ET AL: "The Concept and the Epidemiology of Diabetic Nephropathy Have Changed in Recent Years", J CLIN MED, vol. 4, 2015, pages 1207 - 1216
MCINNES I.B, ET AL: "Cytokines in the pathogenesis of rheumatoid arthritis", NAT REV IMMUNOL, vol. 7, 2007, pages 429 - 442
MUYDERMAN H. ET AL: "Mitochondrial dysfunction in amyotrophic lateral sclerosis - a valid pharmacological target?", BR J PHARMACOL, vol. 171, 2014, pages 2191 - 2205
NAKAMURA Y. ET AL: "A formal anti-Markovnikov hydroamination of allylic alcohols via tandem oxidation/1, 4-conjugate addition/1,2-reduction using a Ru catalyst", CHEM COMMUN, vol. 51, 2015, pages 7459 - 7462
NAZIROGLU M. ET AL: "Role of oxidative stress and Ca(2)(+) signaling on molecular pathways of neuropathic pain in diabetes: focus on TRP channels", NEUROCHEM RES, vol. 37, 2012, pages 2065 - 2075, XP035111060, DOI: doi:10.1007/s11064-012-0850-x
NICKOLOFF B.J. ET AL: "Immunopathogenesis of psoriasis", CLIN REV ALLERGY IMMUNOL, vol. 33, 2007, pages 45 - 56, XP055054377, DOI: doi:10.1007/s12016-007-0039-2
NORMAN J: "The role of cytokines in the pathogenesis of acute pancreatitis", AM J SURG, vol. 175, 1998, pages 76 - 83
OLSSON B. ET AL.: "Microglial markers are elevated in the prodromal phase of Alzheimer's disease and vascular dementia", J ALZHEIMERS DIS, vol. 33, 2013, pages 45 - 53
OVERWATER I.E. ET AL: "Epilepsy in children with tuberous sclerosis complex: Chance of remission and response to antiepileptic drugs", EPILEPSIA, vol. 56, 2015, pages 1239 - 1245
PARACATU L.C. ET AL.: "Alkyl caffeates as anti-Helicobacter pylori and scavenger of oxidants produced by neutrophils", MED CHEM, vol. 10, 2014, pages 74 - 80
PODOLSKY DK: "Inflammatory bowel disease", N ENGL J MED, vol. 347, 2002, pages 417 - 429
PRADHAN S. ET AL: "Commentary: Progressive inflammation as a contributing factor to early development of Parkinson's disease", EXP NEUROL, vol. 241, 2013, pages 148 - 155
RAASCHOU P. ET AL: "Rheumatoid arthritis, anti-tumour necrosis factor therapy, and risk of malignant melanoma: nationwide population based prospective cohort study from Sweden", BMJ, vol. 346, 2013, pages FL939
REICHERT F. ET AL: "Complement-receptor-3 and scavenger-receptor-AI/II mediated myelin phagocytosis in microglia and macrophages", NEUROBIOL DIS, vol. 12, 2003, pages 65 - 72
REUTER S. ET AL: "Oxidative stress, inflammation, and cancer: how are they linked?", FREE RADIC BIOL MED, vol. 49, 2010, pages 1603 - 1616, XP027452772, DOI: doi:10.1016/j.freeradbiomed.2010.09.006
RODRIGO R. ET AL.: "Oxidative stress and pathophysiology of ischemic stroke: novel therapeutic opportunities", CNS NEUROL DISORD DRUG TARGETS, vol. 12, 2013, pages 698 - 714
RONGIOLETTI F. ET AL: "Inflammatory/infectious cutaneous side effects of biological drugs in patients with psoriasis: a general review with personal data", G ITAL DERMATOL VENEREOL, vol. 149, 2014, pages 311 - 316
ROY S. ET AL: "Synthesis of 7-keto-Go6976 (ICP-103", SYNTH COMMUN, vol. 35, 2005, pages 595 - 601
ROY S. ET AL: "Synthesis of N-alkyl substituted bioactive indolocarbazoles related to G66976", TETRAHEDRON, vol. 62, 2006, pages 7838 - 7845, XP025002502, DOI: doi:10.1016/j.tet.2006.05.049
RUBINO M.T. ET AL: "Identification of novel matrix metalloproteinase inhibitors by screening of phenol fragments library", ARCH PHARM CHEM.LIFE SCI., vol. 344, 2011, pages 557 - 563
SCHRAG M. ET AL.: "Oxidative stress in blood in Alzheimer's disease and mild cognitive impairment: a meta-analysis", NEUROBIOL DIS, vol. 59, 2013, pages 100 - 110, XP028713008, DOI: doi:10.1016/j.nbd.2013.07.005
SCHMID R.M. ET AL: "Cytokines in acute pancreatitis: New pathophysiological concepts evolve", EUR J GASTROENTEROL HEPATOL, 1999, pages 125 - 127
SCHULZ E. ET AL: "Oxidative stress, antioxidants, and endothelial function", CURR MED CHEM, vol. 11, 2004, pages 1093 - 1104
SEROR R. ET AL.: "Pattern of demyelination occurring during anti-TNF-alpha therapy: a French national survey", RHEUMATOLOGY (OXFORD, vol. 52, 2013, pages 868 - 874
SHAPIRO S.L. ET AL: "Aminoalkylamides and Oxazolidinediones", J AM CHEM SOC, vol. 81, 1959, pages 3083 - 3088, XP002285705, DOI: doi:10.1021/ja01521a043
SHIROTA O. ET AL: "Concise large-scale synthesis of psilocin and psilocybin, principal hallucinogenic constituents of ''magic mushroom", J NAT PROD, vol. 66, 2003, pages 885 - 887
SMITH K.J. ET AL: "Demyelination: the role of reactive oxygen and nitrogen species", BRAIN PATHOL, vol. 9, 1999, pages 69 - 92
SMITH M.A. ET AL.: "Chronological primacy of oxidative stress in Alzheimer disease", NEUROBIOL AGING, vol. 26, 2005, pages 579 - 580, XP004748491, DOI: doi:10.1016/j.neurobiolaging.2004.09.021
SONG C; HALBREICH U; HAN C; LEONARD BE; LUO H: "Imbalance between pro- and anti-inflammatory cytokines, and between Thl and Th2 cytokines in depressed patients: the effect of electroacupuncture or fluoxetine treatment", PHARMACOPSYCHIATRY, vol. 42, 2009, pages 182 - 188
STEINMAN L, ET AL: "How to successfully apply animal studies in experimental allergic encephalomyelitis to research on multiple sclerosis", ANN NEUROL, vol. 60, 2006, pages 12 - 21
STREETZ K. AL.: "Tumor necrosis factor alpha in the pathogenesis of human and murine fulminant hepatic failure", GASTROENTEROLOGY, vol. 119, 2000, pages 446 - 460, XP029479366, DOI: doi:10.1053/gast.2000.9364
SU K.H. ET AL: "Review of experimental animal models of acute pancreatitis", HPB, vol. 8, 2006, pages 264 - 286
TEE A.R. ET AL: "Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling", PROC NATL ACAD SCI USA, vol. 99, 2002, pages 13571 - 13576
TEE A.R. ET AL: "Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb", CURRENT BIOL, vol. 13, 2003, pages 1259 - 1268
ULICH T.R. ET AL.: "Intratracheal injection of LPS and cytokines. V. LPS induces expression of LIF and LIF inhibits acute inflammation", AM J PHYSIOL, vol. 267, 1994, pages L442 - 446
UNDINE E. ET AL, MOLECULAR MECHANISMS OF DEPRESSION: PERSPECTIVES ON NEW TREATMENT STRATEGIES CELL PHYSIOL BIOCHEM, vol. 31, 2013, pages 761 - 777
VAN ACKER GJD ET AL: "Cause-effect relationships between zymogen activation and other early events in secretagogue-induced acute pancreatitis", AM. J. PHYSIOL. GASTROINTES. LIVER PHYSIOL, vol. 292, 2007, pages G1738 - G1746
VAN DARTEL SA ET AL.: "Predictors for the 5-year risk of serious infections in patients with rheumatoid arthritis treated with anti-tumour necrosis factor therapy: a cohort study in the Dutch Rheumatoid Arthritis Monitoring (DREAM) registry", RHEUMATOLOGY (OXFORD, vol. 52, 2013, pages 1052 - 1057
VAN DEVENTER SJ.: "Tumour necrosis factor and Crohn's disease", GUT, vol. 40, 1997, pages 443 - 448
WATTERS J.J. ET AL: "A differential role for the mitogen-activated protein kinases in lipopolysaccharide signaling: the MEK/ERK pathway is not essential for nitric oxide and interleukin lbeta production", J BIOL CHEM, vol. 277, 2002, pages 9077 - 9087
WITTIG G. ET AL: "Benzo-cycloheptatrien, Benzo-cyclooctatrien und Benzo-cyclooctatetraen: VI. Mitteilung uber neuartige Synthesen von Cyclopolyenen", JUSTUS LIEBIGS ANN CHEM., vol. 619, 1958, pages 10 - 27
YANOVSKY I. ET AL: "Carbamate Derivatives of Indolines as Cholinesterase Inhibitors and Antioxidants for the Treatment of Alzheimer's Disease", J MED CHEM, vol. 55, 2012, pages 10700 - 10715, XP055071815, DOI: doi:10.1021/jm301411g
YAO C-H ET AL: "Discovery of novel N-(3-d-xylosylindole derivatives as sodium-dependent glucose cotransporter 2 (SGLT2) inhibitors for the management of hyperglycemia in diabetes", J MED CHEM, vol. 54, 2010, pages 166 - 178, XP009161759, DOI: doi:10.1021/jm101072y
YASUNO F. Y ET AL.: "Increased binding of peripheral benzodiazepine receptor in mild cognitive impairment-dementia converters measured by positron emission tomography with [(1)(1)C]DAA1106", PSYCHIATRY RES, vol. 203, 2012, pages 67 - 74, XP028939333, DOI: doi:10.1016/j.pscychresns.2011.08.013
YEOM C-E ET AL: "1,8-Diazabicyclo[5.4.0]undec-7-ene (DBU)-promoted efficient and versatile aza-Michael addition", TETRAHEDRON, vol. 63, 2007, pages 904 - 909, XP005805696, DOI: doi:10.1016/j.tet.2006.11.037
BERGE ET AL, J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
Attorney, Agent or Firm:
PEARL, Zeev (IL)
Download PDF:
Claims:
CLAIMS:

1. A compound of general formula (I), including any stereoisomer and salt thereof:

wherein

Ri is selected from Ci-Cio alkoxy, aryloxy, benzyloxy, hydroxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -OH, - C(=0)OH, and -C(=0)NR3R4;

R3 and R4 are each independently selected from H, straight or branched C1-C10 alkyl and aryl.

2. A compound of general formula (Γ), including any stereoisomer and salt thereof:

wherein

Ri is selected from C1-C10 alkoxy, aryloxy, benzyloxy, hydroxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl; R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one of -NH2, -NHR5, -NR6R7;

R5, R6, and R7 are each independently selected from straight or branched C1-C10 alkyl and aryl;

provided that when R2 is C2 alkyl it is substituted by at least one of - NHRs, -NR6R7; and

provided that when R2 is a straight C3-C8 alkyl it is substituted by at least one of -NH2, -NHR5 and -NR6R7 wherein R5, R6, and R7 are each independently selected from branched C3-C10 alkyl or an aryl.

3. A compound according to claims 1 or 2, wherein Ri is selected from C1-C10 alkoxy, aryloxy, benzyloxy, hydroxy and halogen.

4. A compound according to claims 1 or 2, wherein Ri is -0(Ci-Cs alkyl).

5. A compound according to claims 1 or 2, wherein Ri is a halogen.

6. A compound according to claims 1 or 2, wherein Ri is benzyloxy.

7. A compound according to claims 1 or 2, wherein Ri is hydroxy.

8. A compound according to any one of claims 1 to 7, wherein Ri is substituted at position 4.

9. A compound according to any one of claims 1 to 7, wherein Ri is substituted at position 5.

10. A compound according to any one of claims 1 to 7, wherein Ri is substituted at position 6.

11. A compound according to any one of claims 1 to 7, wherein Ri is substituted at position 7.

12. A compound according to any one of claims 1 and 3 to 11, wherein R2 is a straight or branched C2-C8 alkyl substituted by at least one group selected from - OH, -C(=0)0(Ci-C5alkyl), -C(=0)OH, and -C(=0)NR3R4.

13. A compound according to any one of claims 1 and 3 to 11, wherein R2 is a straight or branched C2-C8 alkyl substituted by at least one -OH.

14. A compound according to any one of claims 1 and 3 to 11, wherein R2 is a straight or branched C2-C8 alkyl substituted by at least one

15. A compound according to claim 2, wherein R2 is a straight or branched C3-C8 alkyl substituted by at least one -NH2.

16. - A compound according to claim 2, wherein R2 is a straight or branched C3-C8 alkyl substituted by at least one -NHR5.

17. A compound according to claim 2, wherein R2 is a straight or branched C3-C8 alkyl substituted by at least one -NR6R7.

18. A compound according to any one of claims 1 and 3 to 14, wherein R2 is a straight or branched C2-C8 alkyl substituted by at least one wherein R3 and R4 are each independently selected from H, straight or branched C1-C10 alkyl and aryl.

19. A compound according to any one of claims 1 to 18, being a pharmaceutically acceptable salt thereof.

20. A compound according to any one of claims 1 to 18, for use as a medicament.

21. A compound according to any one of claims 1 to 18, for use in the reduction of at least one condition selected from oxidative stress, release of NO and release of proinflammatory cytokine.

22. A compound according to any one of claims 1 to 18, for use in the inhibition of at least one of oxidative stress, inflammation.

23. A compound according to any one of claims 1 to 18, for use in the prevention, treatment or slowing the progression of a neurodegenerative disease, disorder or condition including any symptoms thereof.

24. A compound according to any one of claims 1 to 18, for use in the prevention, treatment or slowing the progression of an inflammatory disease, disorder or condition including any symptoms thereof.

25. A compound according to any one of claims 1 to 18, for use in the treatment, prevention or slowing the progression of a disease, disorder, condition or symptom selected from at least one of Alzheimer's disease, Parkinson's disease, depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, mild cognitive impairment, ulcerative colitis, Crohn's disease, pancreatitis, rheumatoid arthritis, diabetes, cardiac failure, chronic liver disease, chronic lung disease, meningitis, infective brain disease, complex regional pain syndrome (CRPS), tuberous sclerosis, psoriasis and any combinations thereof.

26. A composition comprising at least one compound of any one of claims 1 to 15.

27. Use of a compound according to any one of claims 1 to 18, for the preparation of a medicament.

28. Use of a compound according to any one of claims 1 to 18, for the preparation of a medicament for the treatment of a disease, disorder, condition or symptom associated with the inhibition of at least one of oxidative stress, inflammation.

29. Use of a compound according to any one of claims 1 to 18, for the preparation of a medicament for the reduction of at least one condition selected from oxidative stress, release of NO and release of pro-inflammatory cytokine.

30. Use according to claims 28 or 29, wherein said disease, disorder, condition or symptom is selected from at least one of Alzheimer's disease, Parkinson's disease, depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, mild cognitive impairment, ulcerative colitis, Crohn's disease, pancreatitis, rheumatoid arthritis, diabetes, cardiac failure, chronic liver disease, chronic lung disease, meningitis, infective brain disease, complex regional pain syndrome (CRPS), tuberous sclerosis, psoriasis and any combinations thereof.

31. Use of a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein Ri is selected from H, OH, Ci-Cio alkoxy, aryloxy, benzyloxy, hydroxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched C2-C6 alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)OH, - C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the reduction of at least one condition selected from oxidative stress, release of NO and release of pro-inflammatory cytokine.

32. Use of a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein

Ri is selected from H, OH, C1-C10 alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)0(Ci-Csalkyl), -C(=0)OH,- C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5, R6 and R?are each independently selected from H, straight or branched C1-C10 alkyl and aryl; for the preparation of a medicament for the inhibition of at least one of oxidative stress and inflammation.

33. Use of a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein

Ri is selected from H, OH, Ci-Cio alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)OH,- C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the prevention, treatment or slowing the progression of a neurodegenerative disease, disorder or condition including any symptoms thereof.

34. Use of a compound of general formula (I"), including any stereoisomer and salt thereof:

4

(Γ')

wherein

Ri is selected from H, OH, Ci-Cio alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)0(Ci-Csalkyl),-C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the prevention, treatment or slowing the progression of an inflammatory disease, disorder or condition including any symptoms thereof.

35. Use according to any one of claims 32 to 34, wherein said disease, disorder, condition or symptom selected from at least one of Alzheimer's disease, Parkinson's disease, depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, mild cognitive impairment, ulcerative colitis, Crohn's disease, pancreatitis, rheumatoid arthritis, diabetes, cardiac failure, chronic liver disease, chronic lung disease, meningitis, infective brain disease, complex regional pain syndrome (CRPS), tuberous sclerosis, psoriasis and any combinations thereof.

36. A compound according to claims 1 or 2, selected from the following:

! l

for the preparation of a medicament for the prevention, treatment or slowing the progression of a disease, disorder or condition including any symptoms thereof, selected from inflammatory disease, neurodegenerative disease, disease associated with at least one of oxidative stress, an immune response, release of NO and release of proinflammatory cytokine.

38. A compound according to any one of claims 1 to 18, for use in the immunomodulation of a condition, disease or disorder associated with an immune response.

39. A pharmaceutical composition comprising a compound of general formula (II), including any stereoisomer and salt thereof:

wherein

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl; wherein R5 is different than H; and at least one of R3 and R4 or at least one of R6 and R7 is different than H.

40. The pharmaceutical composition of claim 39, R2 is a straight or branched C2-C8 alkyl substituted by at least one group selected from -C(=0)NR3R4, -NHR5 and -NR6R7; R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched Ci- C10 alkyl wherein R5 is different than H and at least one of R3 and R4 or at least one of R6 and R7 is different than H.

41. The pharmaceutical composition of claim 39, wherein R2 is a straight or branched C2-C8 alkyl substituted by at least one -NHR5 or -NR6R7; wherein R5, R6, and R7 are each independently selected from H, straight or branched C1-C10 alkyl, and wherein R5 is different than H and at least one of R3 and R4 is different than H.

42. The pharmaceutical composition of claim 39, wherein R2 is a straight or branched C2-C8 alkyl substituted by at least one -C(=0)NR3R4; wherein R3 and R4 are each independently selected from H, straight or branched C1-C10 alkyl and wherein at least one of R3 and R4 is different than H.

43. The pharmaceutical composition of claims 39, being selected from:

44. A use of a compound of general formula (ΙΓ), including any stereoisomer and salt thereof:

wherein

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -OH, - C(=0)OH , -C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5 R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the prevention, treatment or slowing the progression of a disease, disorder or condition including any symptoms thereof, selected from inflammatory disease, neurodegenerative disease, disease associated with at least one of oxidative stress, an immune response, release of NO and release of proinflammatory cytokine.

45. The use of claim 44, wherein R2 is a straight or branched C2-C8 alkyl substituted with -C(=0)NR3R4, -NHR5 and -NR6R7.

46. The use of claim 44, wherein R2 is substituted with -NHR5 and -NR6R7.

47. The use of claim 44, wherein Ra is substituted with -C(=0)NR3R4-

48. The use of claim 44, wherein R2 is substituted with OH.

49. The use of claim 44, wherein R2 is substituted with

50. The use of claim 44, wherein said compound is selected from:

51. A pharmaceutical composition comprising a compound of general formula (III), including any stereoisomer and salt thereof:

wherein

R2 is selected from straight or branched C3-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)NR3R4, -NHR5 and -NR6R7;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl.

52. The composition of claim 51, wherein the compound of formula (III) is selected from:

53. Use of a compound of general formula (IV), including any stereoisomer and salt thereof:

wherein

Ri is selected from H, OH, C1-C10 alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)NR3R4, -NHRs and -NR6R7;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the prevention, treatment or slowing the progression of a disease, disorder or condition including any symptoms thereof, selected from inflammatory disease, neurodegenerative disease, disease associated with at least one of oxidative stress, an immune response, release of NO and release of proinflammatory cytokine.

Description:
INDOLINE DERIVATIVES, COMPOSITIONS COMPRISING THEM AND

USES THEREOF

TECHNOLOGICAL FIELD

This invention relates to indoline derivatives and salts thereof, compositions comprising them and uses thereof for the treatment of diseases and disorders.

BACKGROUND ART

References considered that are relevant as background to the presently disclosed subject matter are listed below:

Adler S, Kolev M, Varisco PA, Tham M, von Gunten M, Tappeiner C et al. (2013). Induction of severe systemic lupus erythematosus by TNF blockade and response to anti-IL-6 strategy. J Allergy Clin Immunol 131 : 1235-1237, 1237 el231.

Allez M, Karmiris K, Louis E, Van Assche G, Ben-Horin S, Klein A, et al. (2010) Report of the ECCO pathogenesis workshop on anti-TNF therapy failures in inflammatory bowel diseases: definitions, frequency and pharmacological aspects. J Crohns Colitis 4(4):355-366.

Amundsen LH, Nelson LS. (1951). Reduction of nitriles to primary amines with lithium aluminum hydride. J Am Chem Soc 73: 242-244.

Astil B, Boekelheide V. (1958). Synthesis of 5-Ketolilolidine. J Org Chem 23: 316-318.

Baumann H, Gauldie J. (1994). The acute phase response. Immunol Today 15:

74-80.

Ben-Horin S, Chowers Y. (2014). Tailoring anti-TNF therapy in IBD: drug levels and disease activity. Nat Rev Gastroenterol Hepatol. l l(4):243-255.

Berney T, Gasche Y, Robert J, Jenny A, Mensi N, Grau G, Vermeulen B, and Morel P (1999). Serum profiles of interleukin-6, interleukin-8, and inter leukin- 10 in patients with severe and mild acute pancreatitis. Pancreas 18:371-377

Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. (2006). Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA 295: 2275-2285.

Cornu M, Albert V, Hall MN. (2013). mTOR in aging, metabolism, and cancer, Curr Op Gen Dev 23:53-62.

DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR et al. (2013). B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci USA 110: 5133-5138.

DellaGioia N, Devine L, Pittman B, Hannestad J. (2013). Bupropion pre- treatment of endotoxin-induced depressive symptoms. Brain Behav Immun 31 : 197-204.

Dhir A, Kulkarni SK. (2007). Involvement of L-arginine-nitric oxide-cyclic guanosine monophosphate pathway in the antidepressant-like effect of venlafaxine in mice. Prog Neuropsychopharmacol Biol Psychiatry 31 : 921-925.

Dias V, Junn E, Mouradian MM. (2013). The role of oxidative stress in Parkinson's disease. J Parkinson's Dis 3: 461-491.

Dinarello CA. (1997). Pro-inflammatory and anti-inflammatory cytokines as mediators in the pathogenesis of septic shock. Chest 112: 321S-329S.

Gonzalez-Juanatey C, Vazquez-Rodriguez TR, Miranda-Filloy JA, Gomez- Acebo I, Testa A, Garcia-Porrua C et al. (2012). Anti-TNF-alpha-adalimumab therapy is associated with persistent improvement of endothelial function without progression of carotid intima-media wall thickness in patients with rheumatoid arthritis refractory to conventional therapy. Mediators Inflamm 2012: 674265.

Hajjar DP, Gotto AM, Jr. (2013). Biological relevance of inflammation and oxidative stress in the pathogenesis of arterial diseases. Am J Pathol 182: 1474-1481.

Irony- Tur-Sinai M, Grigoriadis N, Lourbopoulos A, Pinto-Maaravi F, Abramsky O, Brenner T. (2006). Amelioration of autoimmune neuroinflammation by recombinant human alpha-fetoprotein. Exp Neurol 198: 136-144.

Kanasaki K, Taduri G, Koya D. (2013). Diabetic nephropathy: the role of inflammation in fibroblast activation and kidney fibrosis. Front Endocrinol (Lausanne). Feb 6; 4: 7-15.

Levitt JE, Matthay MA. (2006). Treatment of acute lung injury: historical perspective and potential future therapies. Semin Respir Crit Care Med 27: 426-437. Maes M. (1999). Major depression and activation of the inflammatory response system. Adv Exp Med Biol 461 : 25-46.

Maes M. (2013). Inflammatory and oxidative and nitrosative stress cascades as new drug targets in myalgic encephalomyelitis and chronic fatigue syndrome. Mod Trends Pharmacopsychiatry 28: 162-174.

Magro F, Portela F. (2010). Management of inflammatory bowel disease with infliximab and other anti-tumor necrosis factor alpha therapies. BioDrugs 24 Suppl 1: 3- 14.

Mandell BF, Sobell JM. (2014). The role of TNF inhibitors in psoriatic disease. Semin Cutan Med Surg 33: S64-68.

Mangialasche F, Polidori MC, Monastero R, Ercolani S, Camarda C, Cecchetti R et al. (2009). Biomarkers of oxidative and nitrosative damage in Alzheimer's disease and mild cognitive impairment. Ageing Res Rev 8: 285-305.

Martinez-Castelao A, Navarro-Gonzalez JF, Gorriz JL, de Alvaro F (2015). The Concept and the Epidemiology of Diabetic Nephropathy Have Changed in Recent Years. J Clin Med 4: 1207-1216

Mclnnes IB, Schett G. (2007). Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol 7: 429-442.

Muyderman H, Chen T. (2014). Mitochondrial dysfunction in amyotrophic lateral sclerosis - a valid pharmacological target? Br J Pharmacol 171 : 2191-2205.

Nakamura Y, Ohta T, Oe Y. (2015). A formal anti-Markovnikov hydroamination of allylic alcohols via tandem oxidation/1, 4-conjugate addition/1, 2-reduction using a Ru catalyst. Chem Commun 51 : 7459-7462.

Naziroglu M, Dikici DM, Dursun S. (2012). Role of oxidative stress and Ca(2)(+) signaling on molecular pathways of neuropathic pain in diabetes: focus on TRP channels. Neurochem Res 37: 2065-2075.

Nickoloff BJ, Qin JZ, Nestle FO. (2007). Immunopathogenesis of psoriasis. Clin Rev Allergy Immunol 33: 45-56.

Norman J (1998). The role of cytokines in the pathogenesis of acute pancreatitis. Am J Surg 175:76-83.

Olsson B, Hertze J, Lautner R, Zetterberg H, Nagga K, Hoglund K et al. (2013). Microglial markers are elevated in the prodromal phase of Alzheimer's disease and vascular dementia. J Alzheimers Dis 33: 45-53. Overwater IE, Bindels-de Heus K, Rietman AB, Ten Hoopen LW, Vergouwe Y, Moll HA, de Wit MC. (2015) Epilepsy in children with tuberous sclerosis complex: Chance of remission and response to antiepileptic drugs, Epilepsia, 56: 1239-1245.

Paracatu LC, Bonacorsi C, de Farias CM, Nazare AC, Petronio MS, Regasini LO et al. (2014). Alkyl caffeates as anti-Helicobacter pylori and scavenger of oxidants produced by neutrophils. Med Chem 10: 74-80.

Petrovna, VT, Vladimirovna VD, Nikolaevich OS. (2010). Method for synthesis of l-(aminoalkyl)indolines. Patent RU 2387640.

Podolsky DK. (2002). Inflammatory bowel disease. N Engl J Med 347: 417-429.

Pradhan S, Andreasson K. (2013). Commentary: Progressive inflammation as a contributing factor to early development of Parkinson's disease. Exp Neurol 241 : 148- 155.

Raaschou P, Simard JF, Holmqvist M, Askling J, Group AS. (2013). Rheumatoid arthritis, anti-tumour necrosis factor therapy, and risk of malignant melanoma: nationwide population based prospective cohort study from Sweden. BMJ 346: fl939.

Reichert F, Rotshenker S. (2003). Complement-receptor-3 and scavenger- receptor- AI/II mediated myelin phagocytosis in microglia and macrophages. Neurobiol Dis 12: 65-72.

Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. (2010). Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49: 1603-1616.

Rodrigo R, Fernandez-Gajardo R, Gutierrez R, Matamala JM, Carrasco R, Miranda-Merchak A et al. (2013). Oxidative stress and pathophysiology of ischemic stroke: novel therapeutic opportunities. CNS Neurol Disord Drug Targets 12: 698-714.

Rongioletti F, Burlando M, Parodi A. (2014). Inflammatory/infectious cutaneous side effects of biological drugs in patients with psoriasis: a general review with personal data. G Ital Dermatol Venereol 149: 311-316.

Roy S, Eastman A, Gribble GW. (2005). Synthesis of 7-keto-Go6976 (ICP- 103). Synth Commun, 35: 595-601.

Roy S, Eastman A, Gribble GW. (2006). Synthesis of N-alkyl substituted bioactive indolocarbazoles related to Go6976. Tetrahedron, 62: 7838-7845.

Rubino MT, Maggi D, Laghezza A, Loiodice F, Tortorella P. (2011). Identification of novel matrix metalloproteinase inhibitors by screening of phenol fragments library. Arch Pharm Chem.Life Sci. 344: 557-563 Schrag M, Mueller C, Zabel M, Crofton A, Kirsch WM, Ghribi O et al. (2013). Oxidative stress in blood in Alzheimer's disease and mild cognitive impairment: a metaanalysis. Neurobiol Dis 59: 100-110.

Schmid RM and Adler G (1999). Cytokines in acute pancreatitis: New pathophysiological concepts evolve. Eur J Gastroenterol Hepatol 125-127.

Schulz E, Anter E, Keaney JF, Jr. (2004). Oxidative stress, antioxidants, and endothelial function. Curr Med Chem 11: 1093-1104.

Seror R, Richez C, Sordet C, Rist S, Gossec L, Direz G et al. (2013). Pattern of demyelination occurring during anti-TNF-alpha therapy: a French national survey. Rheumatology (Oxford) 52: 868-874.

Shapiro SL, Rose IM, Freedman L. (1959). Aminoalkylamides and Oxazolidinediones. J Am Chem Soc 81: 3083-3088.

Shirota O, Hakamata W, Goda Y. (2003) Concise large-scale synthesis of psilocin and psilocybin, principal hallucinogenic constituents of "magic mushroom". J Nat Prod 66: 885-887.

Smith KJ, Kapoor R, Felts PA. (1999). Demyelination: the role of reactive oxygen and nitrogen species. Brain Pathol 9: 69-92.

Smith MA, Nunomura A, Lee HG, Zhu X, Moreira PI, Avila J et al. (2005). Chronological primacy of oxidative stress in Alzheimer disease. Neurobiol Aging 26: 579-580.

Song C, Halbreich U, Han C, Leonard BE, Luo H. (2009). Imbalance between pro- and anti-inflammatory cytokines, and between Thl and Th2 cytokines in depressed patients: the effect of electroacupuncture or fluoxetine treatment. Pharmacopsychiatry 42: 182-188.

Steinman L, Zamvil SS. (2006). How to successfully apply animal studies in experimental allergic encephalomyelitis to research on multiple sclerosis. Ann Neurol 60: 12-21.

Streetz K, Leifeld L (2000). Grundmann D, Ramakers J, Eckert K, Spengler U, et al. Tumor necrosis factor alpha in the pathogenesis of human and murine fulminant hepatic failure. Gastroenterology 119:446-460.

Su KH, Cuthbertson C. Christophi C. (2006) Review of experimental animal models of acute pancreatitis. HPB. 8:264-286. Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC, Blenis J: (2002). Tuberous sclerosis complex- 1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling, Proc Natl Acad Sci USA, 99: 13571-13576.

Tee AR, Manning BD, Roux PP, Cantley LC, Blenis J. (2003). Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb, Current Biol 13: 1259-1268.

Ulich TR, Fann MJ, Patterson PH, Williams JH, Samal B, Del Castillo J et al. (1994). Intratracheal injection of LPS and cytokines. V. LPS induces expression of LIF and LIF inhibits acute inflammation. Am J Physiol 267: L442-446.

Undine E. Borgwardt L.S. (2013) Molecular Mechanisms of Depression: Perspectives on New Treatment Strategies Cell Physiol Biochem 31:761-777.

Van Acker GJD, Weiss E, Steer ML. Perides G. (2007). Cause-effect relationships between zymogen activation and other early events in secretagogue-induced acute pancreatitis. Am. J. Physiol. Gastrointes. Liver Physiol; 292:G1738-G1746.

van Dartel SA, Fransen J, Kievit W, Dutmer EA, Brus HL, Houtman NM et al. (2013). Predictors for the 5-year risk of serious infections in patients with rheumatoid arthritis treated with anti-tumour necrosis factor therapy: a cohort study in the Dutch Rheumatoid Arthritis Monitoring (DREAM) registry. Rheumatology (Oxford) 52: 1052- 1057.

Van Deventer SJ. (1997). Tumour necrosis factor and Crohn's disease. Gut 40: 443-448.

Watters JJ, Sommer JA, Pfeiffer ZA, Prabhu U, Guerra AN, Bertics PJ (2002). A differential role for the mitogen-activated protein kinases in lipopolysaccharide signaling: the MEK/ERK pathway is not essential for nitric oxide and interleukin lbeta production. J Biol Chem 277: 9077-9087.

Wittig G, Eggers H, Duffner P. (1958). Benzo-cycloheptatrien, Benzo- cyclooctatrien und Benzo-cyclooctatetraen: VI. Mitteilung uber neuartige Synthesen von Cyclopolyenen. Justus Liebigs Ann Chem. 619: 10-27

Yanovsky I, Finkin-Groner E, Zaikin A, Lerman L, Shalom H, Zeeli S, Weill T, Ginsburg I, Nudelman A, Weinstock M. (2012) Carbamate Derivatives of Indolines as Cholinesterase Inhibitors and Antioxidants for the Treatment of Alzheimer's Disease. J Med Chem 55: 10700-10715. Yao C-H, Song J-S, Chen C-T, Yeh T-K, Hung M-S, Chang, C-C, Liu, Y-W, Yuan, M-C, Hsieh, C-J, Huang, C-Y, Wang, M-H, Chiu, C-H, Hsieh, T-C, Wu, S-H, Hsiao, W-C, Chu, K-F, Tsai, C-H, Chao, Y-S, Lee, J-C, et al. (2010). Discovery of novel Ν-β-d-xylosylindole derivatives as sodium-dependent glucose cotransporter 2 (SGLT2) inhibitors for the management of hyperglycemia in diabetes. J Med Chem, 54: 166-178

Yasuno F, Kosaka J, Ota M, Higuchi M, Ito H, Fujimura Y et al. (2012). Increased binding of peripheral benzodiazepine receptor in mild cognitive impairment-dementia converters measured by positron emission tomography with [(1)(1)C]DAA1106. Psychiatry Res 203: 67-74.

Yeom C-E, Kim MJ, Kim BM. (2007). l,8-Diazabicyclo[5.4.0]undec-7-ene (DBU)-promoted efficient and versatile aza-Michael addition. Tetrahedron, 63: 904-909

Acknowledgement of the above references herein is not to be inferred as meaning that these are in any way relevant to the patentability of the presently disclosed subject matter.

BACKGROUND

Inflammation is a normal response to infection and injury and involves the recruitment of immune systems to neutralize invading pathogens, repair injured tissues and promote wound healing. (Baumann and Gauldie, 1994). Inflammation is a normal response to infection and injury and involves the recruitment of immune systems to neutralize invading pathogens, repair injured tissues and promote wound healing. (Baumann and Gauldie, 1994). However, chronic or excessive activation of the immune system is associated with an increase in reactive oxygen species (ROS) (Watters et al., 2002), prolonged activation of inducible nitric oxide synthase (iNOS) and the release of pro-inflammatory cytokines e.g. tumor necrosis factor-ot (TNF-ot), interleukin 6 (IL-6), interleukin 1β (IL-Ιβ) (Watters et al., 2002), all of which may increase susceptibility to infections. ROS and pro-inflammatory cytokines have been implicated in a variety of disorders. They include, septic shock (Dinarello, 1997), acute respiratory distress syndrome (Levitt and Matthay, 2006), which can be caused by gram-negative bacteria (Ulich et al., 1994), demyelinating disorders, multiple sclerosis and Guillain-Barre syndrome (Smith et al., 1999), ulcerative colitis (Podolsky, 2002), Crohn's disease (Van Deventer, 1997), rheumatoid arthritis (Mclnnes and Schett, 2007), diabetes, (DeFuria et al., 2013) and diabetic nephropathy (Kanasaki et al., 2013), atherosclerosis (Hajjar and Gotto, 2013), acute liver failure (Streetz et al, 2000), chronic fatigue syndrome (Maes 2013) neuropathic pain (Nazrroglu et al., 2012), psoriasis (Nickoloff et al., 2007) and cancer (Reuter et al., 2010)

Drugs that block the action of TNF-a have proved to be very effective in the treatment of ulcerative colitis, rheumatoid arthritis (Magro and Portela, 2010; Gonzalez- Juanatey et al., 2012; Sorrentino, 2013) and psoriasis (Mandell and Sobell, 2014). However, their chronic use may increase the risk of serious adverse effects, (Raaschou et al, 2013; Bongartz et al, 2006; van Dartel et al., 2013; Seror et al, 2013; Adler et al., 2013; Rongioletti et al., 2014). Also, approximately 33% of patients either do not respond to them or lose response over time within the first 12 months of therapy (Ben Horin et al., 2011), possibly because of the presence of neutralizing antibodies raised against the biological drug. TNF-a antagonists may also cause allergic reactions like myalgias and arthralgias and increase the incidence of infections and lymphomas (Allez et al., 2010).

Impaired mitochondrial function, the production of ROS and an increase in proinflammatory cytokines play a key role in the aetiology of Alzheimer's disease (AD) (Mangialasche et al., 2009). These pathological changes are also seen in Mild Cognitive Impairment, a prodromal form of AD (Schrag et al., 2013; Olsson et al., 2013; Yasuno et al., 2013), to which it progresses in a significant proportion of subjects. Oxidative stress and pro-inflammatory cytokines may contribute to neurodegeneration in other disorders, such as Parkinson's disease (Dias et al., 2013; Pradhan et al., 2013), ischemic stroke (Rodrigo et al., 2013) and amyotrophic lateral sclerosis (Muyderman et al., 2014), and depression (Undine and Borgwardt (2013). An increase in the activation of iNOS and in the levels of pro-inflammatory cytokines has also been implicated in major depressive disorder (Maes 1999; Song et al., 2009) and may be restored to normal by treatment with antidepressants (Dhir and Kulkarni 2007; DellaGioia et al., 2013). For all these conditions there is therefore a need for therapeutic agents that can reduce the damaging effect of oxidative stress and the excessive release of NO and pro- inflammatory cytokines without blocking their receptors, so as not to prevent potentially beneficial effects of the cytokines in other tissues.

Tuberous sclerosis (TS) is a highly prevalent autosomal recessive genetic disease that occurs in approximately 1:6000 births. It causes multi-organ pathologies primarily in the central nervous system (CNS), and in the form of non-malignant tumours, referred to as hamartomas. The CNS defects of TS patients are manifested in epileptic seizures, developmental delays, intellectual disability and autism. The disease, referred to as tuberous sclerosis complex (TSC), has a wide spectrum of severity, which can vary widely between individuals, even between identical twins. This implicates factors other than the genetic lesions themselves as critical for the propagation of the disease. Two genes have been identified as the cause of TSC, hamartin (a.k.a. TSCl) located on chromosome 9 and tuberin (a.k.a. TSC2) located on chromosome 16. A lesion in one of these genes is sufficient to confer the disease. In 2002, TSC was found to function as a key negative regulator of the mammalian target of rapamycin (mTOR), a serine threonine kinase that controls cell metabolism (Tee et al., 2002). Mechanistically, TSCl and TSC2 form a heterodimer to activate the GTPase activity of Rheb, a G-protein critical for the assembly of mTOR into an active complex (Tee et al., 2003). In the absence of either TSCl or TSC2, mTOR is hyperactive. Because mTOR suppression leads to cessation of growth, the gain-of-function mutations in the signalling elements that control mTOR are found in the majority of tumours to support their growth under suboptimal conditions (Cornu et al., 2013). Since TSC patients have an exaggerated mTOR activity the obvious pharmacological strategy for treatment is the use of mTOR inhibitors, such as rapamycin. Rapamycin does not bind mTOR directly, rather inhibits FKBP12, a peptidyl-prolyl cis- trans isomerase necessary for the assembly of mTORCl. In the presence of rapamycin mTORCl disassembles quickly. The disassembly of mTORC2 requires higher concentrations and a longer exposure time. Rapamycin is used as an immunosuppressant following organ transplantation to prevent acute rejection and in graft versus host disease. Everolimus an analogue of rapamycin with better pharmacokinetic properties has been approved by the FDA for the treatment of adults with non-surgically removable brain tumours called subependymal giant cell astrocytomas (SEGA) associated with TSC. This indication was later extended to children.

A second major complication of TSC is the development of kidney tumours called renal angiomyolipomas, which have been reported in up to 75% of patients with the genetic disorder TSC. Lesions are usually multiple, bilateral, and progressive. Other complications of TSC are lung cysts termed lymphangioleiomyomatosis (LAM), a progressive lung disease that usually strikes women during their childbearing years. mTOR inhibitors have some clinical benefit in this condition but do not fully prevent disease progression. The most prevalent symptom of TSC are epileptic seizures, seen from the first year of life, caused by the abnormalities of neuronal migration, cellular differentiation and excessive cellular proliferation. Seizures have a focal or multifocal origin, are often resistant to antiepileptic drugs and impede neurocognitive development. Vigabatrin has proved to be effective against infantile spasms due to TSC (Overwater et al., 2010) but has many side effects in the CNS and periphery and is teratogenic. The genotype - phenotype correlation in TSC is weak so that specific mutations can have a large spectrum of clinical severities, suggesting the influence of other factors on disease progression. This may include the immune system. The molecules of the present application have a broad range of activities, showing no signs of toxicity (in healthy mice), even at a several- fold higher dose, which enable them to be general immunomodulators for various inflammatory conditions.

Diabetic nephropathy (DN) is the most common complication and leading cause of mortality associated with diabetes (Martinez et al., 2015) and its occurrence is expected to increase with diabetes prevalence. Numerous factors, both environmental and genetic, influence the onset, severity and the rate of progression of DN. DN begins with glomerular and tubular hypertrophy and the thickening of the basement membrane and expansion of the mesangium leading to end-stage glomerular closure and tubulointerstitial fibrosis. Inflammation plays a critical role in the pathogenesis, development and progression of diabetic nephropathy (Kanasaki et al., 2013).

The severity of pancreatitis results from immunologic events subsequent to acinar cell injury, including the activation and recruitment of inflammatory cells, the local and systemic production and/or release of cytokines, and the final transmigration of those activated inflammatory cells across the endothelial barrier into the involved tissue (Berney et al., 1999). Experimental evidence therefore suggests that up-regulation of inflammatory mediators including cytokines, chemokines, adhesion molecules, and inducible nitric oxide has a central role in this pathologic process (Norman, 1998; Schmid and Adler, 1999). In humans and experimental animals, the severity of pancreatitis is characterised by elevation in serum amylase GENERAL DESCRIPTION

The present invention provides a compound of general formula (I), including any stereoisomer and salt thereof:

wherein

Ri is selected from Ci-Cio alkoxy, aryloxy, benzyloxy, hydroxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -OH, -C(=0)0(Ci-C 5 alkyl), -C(=0)OH, and -C(=0)NR 3 R 4 ;

R3 and R4 are each independently selected from H, straight or branched C1-C10 alkyl and aryl.

In another aspect the invention provides a compound of general formula (I), including any stereoisomer and salt thereof:

wherein

Ri is selected from C 1 -C 1 0 alkoxy, aryloxy, benzyloxy, hydroxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl; R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one of -NH2, -NHR5, -NR6R7;

R5, R6, and R7 are each independently selected from straight or branched C1-C10 alkyl and aryl;

provided that when R2 is C2 alkyl it is substituted by at least one of - NHRs, -NR 6 R 7 ; and

provided that when R2 is a straight C3-C8 alkyl it is substituted by at least one of -NH2, -NHR5 and -NR6R7 wherein R5, R6, and R7 are each independently selected from branched C3-C10 alkyl or an aryl.

In some embodiments, Ri is selected from C 1 -C 1 0 alkoxy, aryloxy, benzyloxy, hydroxy and halogen. In other embodiments, Ri is -0(Ci-Cs alkyl). In further embodiments, Ri is a halogen. In other embodiments, Ri is benzyloxy. In other embodiments Ri is a hydroxy.

In yet further embodiments, Ri is substituted at position 4. In other embodiments, Ri is substituted at position 5. In further embodiments, Ri is substituted at position 6. In yet other embodiments, Ri is substituted at position 7.

In some embodiments, R2 is a straight or branched C2-C8 alkyl substituted by at least one group selected from -OH, -C(=0)0(Ci-C 5 alkyl), -C(=0)OHand -C(=0)NR 3 R4. In other embodiments, R2 is a straight or branched C2-C8 alkyl substituted by at least one -OH. In yet further embodiments, R2 is a straight or branched C2-C8 alkyl substituted by at least one -C(=0)0(Ci-Csalkyl). In other embodiments, R2 is a straight or branched C2- C8 alkyl substituted by at least one wherein R3 and R4 are each independently and differently selected from H, straight or branched C 1 -C 1 0 alkyl and aryl In other embodiments R2 is substituted by at least one of -NH2, -NHR5, -NR6R7. In other embodiments R2 is substituted by at least one -NH2. In further embodiments R2 is substituted by at least one -NHR5. In yet further embodiments R2 is substituted by at least one -NR6R7. In some embodiments, a compound of the invention is in a form of a pharmaceutically acceptable salt thereof.

In another aspect the invention provides a compound as disclosed herein, for use as a medicament.

In another one of its aspects the present invention provides a pharmaceutical composition comprising a compound of general formula (I), including any stereoisomer and salt thereof:

wherein

Ri is selected from Ci-Cio alkoxy, aryloxy, benzyloxy, hydroxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -OH, -C(=0)0 (Ci-Csalkyl), -C(=0)OH, -NR3R4 and -C(=0)NR 5 R 6 ;

R3, R4, R5 and R6 are each independently selected from H, straight or branched C1-C10 alkyl and aryl.

In yet another aspect, the invention provides a compound as disclosed herein, for use in immunomodulation of a condition, disease or disorder caused by an immune response.

In yet another aspect, the invention provides a compound as disclosed herein, for use in the reduction of at least one condition selected from oxidative stress, release of NO and release of pro-inflammatory cytokines. In yet another aspect, the invention provides a compound as disclosed herein, for use in the inhibition of at least one of oxidative stress and inflammation.

In another aspect, the invention provides a compound as disclosed herein for use in the prevention, treatment or slowing the progression of a neurodegenerative disease, disorder or condition including any symptoms thereof.

In yet another aspect, the invention provides a compound as disclosed herein for use in the prevention, treatment or slowing the progression of an inflammatory disease, disorder or condition including any symptoms thereof.

In a further aspect, the invention provides a compound as disclosed herein, for use in the treatment, prevention or slowing the progression of a disease, disorder, condition or symptom selected from at least one of Alzheimer's disease, Parkinson's disease, depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, mild cognitive impairment, ulcerative colitis, Crohn's disease, pancreatitis, rheumatoid arthritis, diabetes, cardiac failure, chronic liver disease, chronic lung disease, meningitis, infective brain disease, complex regional pain syndrome (CRPS), tuberous sclerosis, psoriasis, and any combinations thereof.

In a further aspect, the invention provides a compound as disclosed herein, for use in the treatment, prevention or slowing the progression of pancreatitis.

In another one of its aspects, the invention provides a composition comprising a compound as disclosed herein.

In another aspect, the invention provides the use of a compound as disclosed herein, for the preparation of a medicament.

In yet another aspect, the invention provides use of a compound as disclosed herein, for the preparation of an immunomodulator for the treatment of a condition, disease or disorder caused by an immune response. In another aspect, the invention provides the use of a compound as disclosed herein, for the preparation of a medicament for the treatment of a disease, disorder, condition or symptom associated with the inhibition of at least one of oxidative stress and inflammation.

In another aspect, the invention provides the use of a compound as disclosed herein, for the preparation of a medicament for the reduction of at least one condition selected from oxidative stress, release of NO and release of pro-inflammatory cytokines.

In some embodiments, said disease, disorder, condition or symptom is selected from at least one of the following; Alzheimer's disease, Parkinson's disease, depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, mild cognitive impairment, ulcerative colitis, Crohn's disease, pancreatitis, rheumatoid arthritis, diabetes, cardiac failure, chronic and acute liver disease, chronic lung disease, meningitis, infective brain disease, complex regional pain syndrome (CRPS), tuberous sclerosis, psoriasis and any combinations thereof.

In another aspect, the invention provides the use a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein

Ri is selected from H, OH, Ci-Cio alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)OH, - C(=0)NR 3 R 4 , -NHRs and -NR 6 R 7 ;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the reduction of at least one condition selected from oxidative stress, release of NO and release of pro-inflammatory cytokine.

In another aspect, the invention provides the use a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein

Ri is selected from H, OH, C 1 -C 1 0 alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)OH, - C(=0)NR 3 R 4 , -NHRs and -NR 6 R 7 ;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C 1 -C 1 0 alkyl and aryl; for the preparation of a medicament for the inhibition of at least one of oxidative stress, inflammation.

In another aspect, the invention provides the use a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein

Ri is selected from H, OH, Ci-Cio alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)0(Ci-Csalkyl), -C(=0)OH, - C(=0)NR 3 R 4 , -NHRs and -NR 6 R 7 ;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C 1 -C 1 0 alkyl and aryl; for the preparation of a medicament for the prevention, treatment or slowing the progression of a neurodegenerative disease, disorder or condition including any symptoms thereof.

In another aspect, the invention provides the use a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein Ri is selected from H, OH, Ci-Cio alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)OH, - C(=0)NR 3 R 4 , -NHRs and -NR 6 R 7 ;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C 1 -C 1 0 alkyl and aryl; for the preparation of a medicament for the prevention, treatment or slowing the progression of an inflammatory disease, disorder or condition including any symptoms thereof.

In another aspect, the invention provides the use a compound of general formula (I"), including any stereoisomer and salt thereof:

(I")

wherein

Ri is selected from H, OH, C 1 -C 1 0 alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)OH, - C(=0)NR 3 R 4 , -NHRs and -NR 6 R 7 ; R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl; for the preparation of a medicament for immunomodulation of a condition, disorder or disease associated with an immune response.

The invention further provides a pharmaceutical composition comprising a compound of general formula (II), including any stereoisomer and salt thereof:

wherein

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)NR3R4, -NHRs and -NR 6 R 7 ;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl wherein R5 is different than H; and at least one of R3 and R4 or at least one of R6 and R7 is different than H.

In some embodiments of the above aspect, R2 is a straight or branched C2-C8 alkyl substituted by at least one group selected from -C(=0)NR3R4, -NHR5 and -NR6R7; R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl wherein R5 is different than H and at least one of R3 and R4 or at least one of R6 and R7 is different than H.

In further embodiments of the above aspect, R2 is a straight or branched C2-C8 alkyl substituted by at least one -NHR5 or -NR6R7; wherein R5, R6, and R7 are each independently selected from H, straight or branched C1-C10 alkyl, and wherein R5 is different than H and at least one of R3 and R4 is different than H. In other embodiments of the above aspect, R2 is a straight or branched C2-C8 alkyl substituted by at least one -C(=0)NR3R4; wherein R3 and R4 are each independently selected from H, straight or branched C1-C10 alkyl and wherein at least one of R3 and R4 is different than H.

In some embodiments of the above aspect, a compound of formula (II) is:

The invention further provides a use of a compound of general formula (ΙΓ), including any stereoisomer and salt thereof:

wherein

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -OH, - C(=0)OH , -C(=0)NR 3 R 4 , -NHRs and -NR 6 R 7 ;

R3, R4, R5 R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

for the preparation of a medicament for the prevention, treatment or slowing the progression of a disease, disorder or condition including any symptoms thereof, selected from inflammatory disease, neurodegenerative disease, disease associated with at least one of oxidative stress, an immune response, release of NO and release of proinflammatory cytokine.

In some embodiments of the above aspect, R2 is a straight or branched C2-C8 alkyl substituted with -C(=0)NR3R4, -NHR5 and -NR6R7. In some further embodiments R2 is substituted with -NR3R4. In other embodiments R2 is substituted with -NHR5 and -NR6R7. In other embodiments R2 is substituted with In other embodiments, R2 is substituted with OH. In further embodiments, R2 is substituted with -C(=0)0(Ci-C 5 alkyl).

In some embodiments of the above aspect, a compound of formula (II) is selected from:

In a further aspect the invention provides a pharmaceutical composition comprising a compound of general formula (III), including any stereoisomer and salt thereof:

wherein

R2 is selected from straight or branched C3-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)NR3R4, -NHR5 and -NR6R7; R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C1-C10 alkyl and aryl;

provided that when R2 is a C2 alkyl that R3, R4, R5, R6 and R7 are each independently selected from straight or branched C 1 -C 1 0 alkyl and aryl.

In some embodiments, a compound of formula (III) is:

The invention also encompasses the use of a compound of general formula (IV), including any stereoisomer and salt thereof:

wherein

Ri is selected from H, OH, C 1 -C 1 0 alkoxy, aryloxy, benzyloxy, halogen, straight or branched C1-C5 alkyl, straight or branched Ci-Ce alkenyl; straight or branched Ci-Ce alkynyl;

R2 is selected from straight or branched C2-C8 alkyl, straight or branched C2-C8 alkenyl; straight or branched C2-C8 alkynyl; wherein said alkyl, alkenyl and alkynyl are each substituted by at least one group selected from -C(=0)NR3R4, -NHRs and -NR 6 R 7 ;

R3, R4, R5, R6 and R7 are each independently selected from H, straight or branched C 1 -C 1 0 alkyl and aryl; for the preparation of a medicament for the prevention, treatment or slowing the progression of a disease, disorder or condition including any symptoms thereof, selected from inflammatory disease, neurodegenerative disease, disease associated with at least one of oxidative stress, an immune response, release of NO and release of pro-inflammatory cytokines.

In some embodiments, a com ound of the invention is selected from the following:

The invention further provides the use of a compound that is selected from the following:

for the preparation of a medicament for the prevention, treatment or slowing the progression of a disease, disorder or condition including any symptoms thereof, selected from inflammatory disease, neurodegenerative disease, disease associated with at least one of oxidative stress, an immune response, release of NO and release of proinflammatory cytokine.

The present invention also relates to pharmaceutical compositions comprising compounds of the subject invention in admixture with pharmaceutically acceptable auxiliaries, and optionally other therapeutic agents. The auxiliaries must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof. Pharmaceutical compositions include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration or administration via an implant. The compositions may be prepared by any method well known in the art of pharmacy.

Such methods include the step of bringing in association compounds used in the invention or combinations thereof with any auxiliary agent. The auxiliary agent(s), also named accessory ingredient(s), include those conventional in the art, such as carriers, fillers, binders, diluents, disintegrants, lubricants, colorants, flavouring agents, antioxidants, and wetting agents.

Pharmaceutical compositions suitable for oral administration may be presented as discrete dosage units such as pills, tablets, dragees or capsules, or as a powder or granules, or as a solution or suspension. The active ingredient may also be presented as a bolus or paste. The compositions can further be processed into a suppository or enema for rectal administration.

The invention further includes a pharmaceutical composition, as hereinbefore described, in combination with packaging material, including instructions for the use of the composition for a use as hereinbefore described.

For parenteral administration, suitable compositions include aqueous and nonaqueous sterile injection. The compositions may be presented in unit-dose or multi-dose containers, for example sealed vials and ampoules, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of sterile liquid carrier, for example water, prior to use. For transdermal administration, e.g. gels, patches or sprays can be contemplated. Compositions or formulations suitable for pulmonary administration e.g. by nasal inhalation include fine dusts or mists which may be generated by means of metered dose pressurized aerosols, nebulisers or insufflators. The exact dose and regimen of administration of the composition will necessarily be dependent upon the therapeutic or nutritional effect to be achieved and may vary with the particular formula, the route of administration, and the age and condition of the individual subject to whom the composition is to be administered.

The invention also includes any salt of a compound of formula (I), including any pharmaceutically acceptable salt, wherein a compound of the invention has a net charge (either positive or negative) and at least one counter ion (having a counter negative or positive charge) is added thereto to form said salt. The phrase "pharmaceutically acceptable salt(s)", as used herein, means those salts of compounds of the invention that are safe and effective for pharmaceutical use in mammals and that possess the desired biological activity. Pharmaceutically acceptable salts include salts of acidic or basic groups present in compounds of the invention. Pharmaceutically acceptable acid addition salts include, but are not limited to, hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzensulfonate, p-toluenesulfonate and pamoate (i.e., l,l'-methylene-bis-(2-hydroxy-3-naphthoate)) salts. Certain compounds of the invention can form pharmaceutically acceptable salts with various amino acids. Suitable base salts include, but are not limited to, aluminum, calcium, lithium, magnesium, potassium, sodium, zinc, and diethanolamine salts. For a review on pharmaceutically acceptable salts see BERGE ET AL., 66 /. PHARM. SCI. 1-19 (1977), incorporated herein by reference.

When referring to "Ci-Cio alkoxy" it should be understood to relate to an RO- moiety wherein R is a straight or branched Ci - Cio alkyl. When referring to "aryloxy" it should be understood to relate to an RO- moiety wherein R is a Cs - C12 aryl (either single or fused). When referring to "benzyloxy" it should be understood to relate to an RO- moiety wherein R is a benzyl. When referring to a "halogen" it should be understood to relate any one of F, CI, Br, I. When referring to "straight or branched C1-C5 alkyl" it should be understood to relate to a hydrocarbon straight or branched chain wherein all carbon atoms are boned to each other and to the hydrogen atoms with a single sigma bond. When referring to "straight or branched Ci-Ce alkenyl" it should be understood to relate to a hydrocarbon straight or branched chain wherein at least two carbon atoms are boned to each other with a double bond. When referring to "straight or branched Ci-Ce alkynyl" it should be understood to relate to a hydrocarbon straight or branched chain wherein at least two carbon atoms are boned to each other with a triple bond.

It is to be understood that the compounds provided herein may contain one or more chiral centres. Such chiral centres may have a configuration of either the (R) or (5) configuration. Compositions provided herein comprising compounds of the invention may be enantiomerically pure (i.e. comprise a single enantiomer or diastereomer of a compound of the invention), or include stereoisomeric mixture of compounds of the invention (i.e. mixtures of enantiomenrs, such as a racemic mixture, or mixtures of diastereomers - such mixtures may be equimolar or non-equimolar).

BRIEF DESCRIPTION OF THE DRAWINGS

In order to better understand the subject matter that is disclosed herein and to exemplify how it may be carried out in practice, embodiments will now be described, by way of non-limiting example only, with reference to the accompanying drawings, in which:

Fig. 1 shows the reduction by compounds in caspase 3 activity induced in macrophages by exposure to H2O2. Data represent the mean ± SD from 4-6 replicates per concentration of each compound. All compounds significantly reduced caspase 3 activity /?<0.01. Significantly different from same concentration of the other compounds * /?<0.05.

Figs. 2A-2B shows the reduction by compounds of the levels of IL-6 in the brain (Fig.2A) and liver (Fig. 2B) of LPS-injected mice. Data represent the mean and SEM from 6-12 mice per dose. Significantly different from LPS + saline * /?<0.05; ** /?<0.01. Significantly different from preceding dose, # /?<0.05.

Figs. 3A-3B shows the reduction by compounds of the levels of TNF-a (Fig. 3A) and IL-6 (Fig. 3B) in plasma of LPS-injected mice. Data represent the mean and SEM from 6-12 mice per dose. Significantly different from LPS + saline * /?<0.05; ** /?<0.01. Significantly different from preceding dose, # /?<0.05. Figs. 4A-4B shows the reduction by compounds of the levels of TNF-a (Fig. 4A) and IL-6 (Fig. 4B) in spleen of LPS-injected mice. Data represent the mean and SEM from 6-12 mice per dose. Significantly different from LPS + saline ** /?<0.01. Significantly different from preceding dose, # /?<0.05.

Figs. 5A-5C shows the reduction by compounds of TNF-a (Fig. 5 A), IL-Ιβ (Fig. 5B) and IL12b (Fig. 5C) mRNA in the brain of LPS-injected mice. Data represent the mean and SEM from 6-12 mice for each group. Significantly different from LPS + saline */?<0.05; ** p<0.0l.

Figs. 6A-6B shows the TSCl deletion causes the development of renal cysts and increase in proliferation in situ. Typical histological appearances of the kidneys of mice stained with Fig. 6A. haematoxylin and eosin (H&E) Fig 6B. immunohistochemistry for Ki67.

Fig. 7 shows how the AN1284 treatment improves the well-being of tamoxifen- induced RERT/TSCl f/f mice. AN1284-treated mice lost much less weight and were more active than the untreated controls.

Figs. 8A-8B shows how AN1284 treatment decreases the numbers of MDSC and T cells in the bone marrow (Fig. 8A) and spleen (Fig. 8B) of tamoxifen-induced RERT/TSCl f/f mice. Significantly different from control mice, * /?<0.05, ** /?<0.01; significantly different from untreated RERT/TSCl f/f mice + tamoxifen, # /?<0.05, ## p<0.0l.

Figs. 9A-9B shows how AN 1284 decreases the elevation of cytokines TNF-a and IL-6 in spleen (Fig. 9A) and liver (Fig. 9B) of RERT/TSCl f/f mice + tamoxifen. Significantly different from control mice **. /?<0.01, significantly different from untreated RERT/TSCl f/f mice + tamoxifen, ## /?<0.01.

Fig. 10 shows how AN 1284 treatment reduces the size of renal cysts and in situ proliferation in TSCl -induced KO mice. Shown are representative slices of the kidney with imunohistochemical staining with Ki67 in three of the five mice. AN 1284 reduced by more than 50% the number of positive nuclei compared to the treated mice.

Figs. 11A-11D shows the reduction of renal damage in type 1 diabetic mice with renal nephropathy-deleted mice treated with AN 1284. Change in body weight during the study (Fig. 11 A), change in total fat mass (Fig. 11B); change in lean body mass (Fig. 11C), degree of hyperglycaemia (Fig. 11D). Figs. 12A-12B shows how AN1284 reduces elevation of albumin and urea in urine of mice with diabetes-induced renal dysfunction. Fig. 12A. Urine albumin-to- creatinine ratio (ACR) Fig. 12B. Serum urea levels. Data represent mean ± SEM from 8- 10 mice per group. Significantly different from vehicle treated control mice * /?<0.05; significantly different from vehicle treated mice with STZ-induced diabetes # /?<0.05.

Figs. 13A-13C shows sections of kidney showing glomerular area. Fig. 13A control mouse; Fig. 13B mouse with STZ induced nephropathy, Fig. 13C mouse with STZ induced diabetes that received AN 1284.

Fig. 14A-14B. Shows reduction by AN1284 of glomerular area and of mesangial expansion in the kidney of mice with diabetes-induced renal dysfunction. Fig. 14A. Quantification of glomerular cross-sectional area. Fig. 14B. Quantification of mesangial expansion. Data represent mean ± SEM from 8-10 mice per group. Significantly different from vehicle treated control mice * /?<0.05; significantly different from vehicle treated mice with STZ-induced diabetes # /?<0.05.

Figs. 15A-15F shows mitigation by AN1284 of diabetes-induced renal injury, inflammation and fibrosis. Diabetic-induced increases in renal mRNA expression levels of: Fig. 15A. lipocalin 2. Fig. 15B. Timpl. Fig. 15C. IP-10. Fibrogenic markers, Fig. 15D collagen I. Fig. 15E collagen III. Fig. 15F. fibronectin 1, were significantly normalised by AN1284 (0.5 mg/kg, sc) treatment. Data represent mean ± SEM from 8- 10 mice per group. Significantly different from vehicle treated control mice * /?<0.05; significantly different from vehicle treated mice with STZ-induced diabetes # /?<0.05.

Figs. 16A-16B shows mitigation of pancreatitis by AN1297. Dose related reduction by AN 1927 of serum amylase (Fig. 16A) and B. pancreatic TNF-a (Fig. 16B) in mice with acute pancreatitis induced by cerulean injections. Resolvin-D (RvE) (1 mg/kg) was used as a positive control. Significantly different from untreated mice *, /?<0.05 **, /?<0.01.

DETAILED DESCRIPTION OF EMBODIMENTS

Experimental Section

General Remarks. ^-NMR, 13 C-NMR spectra were obtained on Bruker Avance- DPX-300, Avance-400, Avance-DMX-600 and Avance-III-700 spectrometers. Chemical shifts are expressed in ppm downfield from Me3Si (TMS) used as internal standard. The values are given in δ scale. The "t" is indicative of a multiplet similar to a triplet with second order characteristics. Mass spectra (MS) were obtained on a Varian Mat 731 spectrometer (CI + = chemical ionization). HRMS were obtained on AutoSpec spectrometer (Water company-UK) (CI + CH4). Electron Spray ionization (ESI) was obtained on a Micromass Q-TOF Micro mass spectrometer (Micromass (Waters) UK). HRMS were obtained on Synapt ESI-Q-TOF (Water company-UK). Progress of the reactions was monitored by TLC on silica gel (Merck, Art. 5554). All the flash chromatographic procedures were carried out on silica gel (Merck, Art. 9385). All moisture sensitive reactions were carried out in flame-dried vessels. Melting points were determined on a Fisher- Johns apparatus. Commercially available compounds were used without further purification. The nomenclature of the compounds was given according to ChemDraw Ultra v. 13 and 14 (CambridgeSoft). The numbering on the chemical structures below is arbitrary and it serves only for spectral analysis. Commercial reagents were used without further purification.

Procedure A: N-Alkylation of indoles or indolines with methyl acrylate (Yeom, et al., 2007). To a stirred solution of an indole or an indoline (0.46 mmol), and methyl acrylate (0.7 mmol) in CH3CN (2 mL) was added DBU (0.23 mmol). The mixture was stirred at 50 °C for 19-24 h, extracted with EtOAc, washed with IN HC1 or IN KHSO4 or saturated aqueous KHSO4, dried over Na2S04 and evaporated.

Procedure B : Reduction of indoles to indolines

Method I: (Yeom et al., 2007). To a solution of an indole (1 mmol) in AcOH (8 mL) at 0 °C, was added NaBBjCN (2 mmol). The mixture was stirred at room temperature from 2-16 h. The residue was extracted with EtOAc and saturated aqueous NaHC03, dried over Na2S04 and evaporated.

Method II: (Yao et al, 2010). A solution of an indole (0.3 mmol) in TFA (4 mL) and Et3SiH (0.8 mmol) was heated to 60 °C and stirred for 4.5 h. The residue was extracted with DCM and saturated aqueous NaHC03, dried over Na2S04 and evaporated.

Method III: (Yanovsky et al., 2012). To a solution of an indole (0.3 mmol) in TFA (20 mL) at 0 °C, was added NaBFLt (1.5 mmol). The mixture was stirred at room temperature from 2-16 h and evaporated. The residue was extracted with EtOAc and saturated aqueous NaHC03, dried over Na2S04 and evaporated.

Procedure C: N-Alkylation of indoles with acrylonitrile. (Roy et al., 2005). To a solution of an indole (17 mmol) in dioxane (20 mL) at 0 °C was added acrylonitrile (26 mmol) followed by the drop wise addition of Triton-B (0.8 mL). The resulting mixture was slowly allowed to warm to room temperature and was stirred for 20 h and was then evaporated. KHSO4 1 N was added dropwise to neutralize the basic catalyst, and the solution was extracted with EtOAc, dried over Na2S04 and evaporated.

Procedure D: N-Alkylation of indoles with N-isopropylacrylamide. (Roy et al., 2006). To a stirred solution of an indole (7 mmol) in dioxane was added N- isopropylacrylamide (7 mmol) and KOH (8.4 mmol). The mixture was stirred at 50 °C from 18-72 h and was then filtered and evaporated.

Procedure E: Reduction of a nitrile (Amundsen et al., 1951). To a solution of a nitrile (n g) in anhydrous diethyl ether (60 mL) at 0 °C was slowly added LAH (n g). The resulting mixture was allowed to warm slowly to room temperature and was further stirred for 24 h. To the mixture was then added water n mL, n mL of 15% NaOH and 3n mL of water. The ether solution was filtered through celite and evaporated.

Procedure F: Reduction of amide or esters (Shirota et al., 2003). To a suspension of L1AIH4 (n g) in anhydrous THF was added drop-wise a solution of an amide (n g) in anhydrous THF. The mixture was refluxed for 24 h for amide reduction or 1 h for ester reduction, cooled to room temperature. The reaction was worked up by addition of water n mL, n mL of 15% NaOH and 3n mL of water. The THF solution was filtered through celite and evaporated. To the residue was added IN of HC1 and the solution was extracted with EtOAc. To the aqueous layer was added saturated Na2C03 until reaching pH = 10 and the solution was extracted with EtOAc. The organic layer was then dried over Na2S04 and evaporated to give the product.

Scheme 1. Synthesis of methyl 3-(indolin-1 -yl)propanoates

5a

a) CH 2 =CHCOOMe/MeCN/DBU/50 °C; b) NaCNBH 3 /AcOH or Et 3 SiH/TFA or NaBH /TFA;

c) HCI (g)/diethyl ether ot p-TSA/ t-butyl methyl ether; d) LAH/THF/HCI Indolines substituted with a methyl propionyl chain at position 1 were prepared from the corresponding indoles 1 or indoline 5a upon treatment with methyl acrylate/DBU. Reduction of the N-alkylated indoles 2a-d,f with NaBH3CN/AcOH. (Yeom et al, 2012), or NaBH 4 /TFA (Yanovsky et al., 2012) or Et 3 SiH/TFA (Yao et al., 2010), gave the corresponding indolines 3a-d,f that were isolated as hydrochlorides 4a-c or p-toluenesulfonate salts 4d,f (Scheme 1). In addition, ester reduction of 3a,b followed by acidification gave the alcohols 6a,b.

The salts of the l-(3-aminopropyl) derivatives 10a,g were prepared from the corresponding indoles la,g upon N-alkylation with acrylonitrile, followed by initial reduction of the indolic to the indolinic system and subsequent reduction of the CN to the corresponding aminomethyl group, and the final products were isolated as p- toluenesulfonate salts 10a,g (Scheme 2).

Scheme 2 . Synthesis of 3-(indolin-1 -yl)propan-1 -amines

10a, g 9a, g

(a) CH 2 =CHCN/Triton B/dioxane, 0 °C -> rt; (b) NaCNBH 3 /AcOH, rt;

(c) LiAIH 4 /diethylether; (d) p-TSA/t-butyl methyl ether

An unexpected reaction took place when an attempt was made to debenzylate 9g under catalytic hydrogenation conditions in a Parr apparatus. Apparently, the hydrogenator contained traces of acetone, which condensed with the amine to give the intermediate imine that underwent further reduction and acidification to give the N- isopropylamino derivative 12g (Scheme 3). An alternative approach to the synthesis of the N-isopropylamino derivatives 12a,b, involved the N-alkylation of indoles 1 with N- isopropylamino acrylamide to give 13a,b, followed by reduction of the indole to the indoline and reduction of the amide to the amine with subsequent acidification (Scheme 4).

Scheme 3. Synthesis of 1 -(3-lsopropylamino)propyl)indolines

(a) H 2 /10% Pd/C/ eOH, 4 atm (and traces of acetone in the hydrogenator);

(b) p-TSA/t-butyl methyl ether;

Scheme 4: Synthesis of 3-(indolin-1 -yl)-N-sopropylpropanamides 15 and 3-(indolin-1 -yl)-N- isopropylpropan-1-amines 12

a) CH 2 =CH-CONH-i-Pr/KOH/Dioxane; b) TFA/NaBH 4 ; c) HCI (g)/Diethyl ether or HCI 3N/EtOAc; d) LAH/THF; e) NaCNBH 3 /AcOH or TFA/NaBH 4 ; f) HCI (g)/Diethyl ether or p-TSA/t-butyl methyl ether A 3-aminopropyl derivative substituted at position 3 of the indoline system 20, was obtained upon amidation of IPA to give amide 17, which underwent two reductive steps and acidification of amine 19 (Scheme 5).

Scheme 5. Synthesis of 3-(indolin-3-yl)propan-1 -amine di-p- toluenesulfonate, 20

20

a) CDI/NH4OH; b) LAH THF; c) Et 3 SH/TFA; d) p-TSA

Preparation Procedures of Specific Compounds Of the Invention

Methyl 3-(lH-Indol-l-yl)propanoate (2a). Compound 2a synthesised from indole by Procedure A, was isolated as a yellow oil in approximately quantitative yield. J H-NMR (400 MHz, CDCb) ppm 5 7.60 (d, / = 8 Hz, 1H), 7.30 (d, / = 8.4 Hz, 1H), 7.19 (t, / = 8.0 Hz, 1H), 7.11-7.06 (m, 2H), 6.45 (d, / = 3.2 Hz), 4.38 (t, / = 6.8 Hz), 3.61 (s, 3H), 2.76 (t, / = 6.8 Hz, 2H); 13 C-NMR (100 MHz, CDCb) ppm δ 171.67, 135.69, 128.78, 127.94, 121.69, 121.11, 119.57, 109.12, 101.65, 51.90, 41.80, 34.76.

Methyl 3-(5-Methoxy-lH-indol-l-yl)propanoate, (2b). Compound 2b synthesised from 5-methoxyindole by Procedure A, was isolated as an orange oil in 94% yield. ¾- NMR (300 MHz, CDCb) ppm δ 7.18 (d, / = 9.0 Hz, 1H), 7.07-7.04 (m, 2H), 6.88-6.83 (m, 1H), 6.38-6.36 (m, 1H), 4.34 (t, / = 6.9 Hz, 2H), 3.80 (s, 3H), 3.60 (s, 3H), 2.74 (t, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 171.66, 154.08, 130.93, 129.06, 128.43, 111.92, 109.82, 102.68, 101.13, 55.79, 51.85, 41.90, 34.78; MS (TOF MS ES + ) m/z 234 (MH+)

Methyl 3-(6-Methyl-lH-indol-l-yl)propanoate, (2c). Compound 2c synthesised from 6-methylindole by Procedure A, was isolated as a yellow oil in 81 % yield. ¾-NMR (400 MHz, CDCb) ppm δ 7.42 (d, / = 8.0 Hz, 1H), 6.99 (s, 1H), 6.87-6.83 (m, 1H), 6.81 (d, / = 3.2 Hz, 1H), 6.32 (d, / = 3.2 Hz, 1H), 4.04 (t, / = 6.8 Hz, 2H), 3.39 (s, 3H), 2.46 (t, / =

6.8 Hz, 2H), 2.40 (s, 3H); 13 C-NMR (100 MHz, CDCb) ppm δ 171.26, 135.79, 130.86, 126.97, 126.32, 121.01 , 120.37, 108.81 , 100.98, 51.17, 41.08, 34.09, 21.46.

Methyl 3-(5-Chloro-lH-indol-l-yl)propanoate, (2d). Compound 2d synthesised from 5-chloroindole by Procedure A, was isolated as an orange oil in 86% yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.51 (d, / = 1.8 Hz, 1H), 7.15-7.03 (m, 3H), 6.34 (d, / = 3.3 Hz, 1H), 4.26 (t, / = 6.9 Hz, 2H), 3.55 (s, 3H), 2.66 (t, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 171.34, 133.94, 129.53, 129.17, 124.97, 121.64, 120.15, 110.08, 101.11, 51.73, 41.67, 34.45; MS (TOF MS ES + ) m/z 238 (MH+), 276 (MK) +

Methyl 3-(6-(Benzyloxy)-lH-indol-l-yl)propanoate, (2f). Compound 2f synthesised from 6-benzyloxyindole by Procedure A, was filtered through a short silica gel column, eluted with EtOAc-hexane (1 :3). Evaporation of the filtrate gave the product as a pink solid, mp 45 °C, in 94% yield. J H-NMR (200 MHz, CDCb) ppm δ 7.55-7.3 (m, 6H), 7.03 (d, / = 3.2 Hz, 2H), 6.93-6.84 (m, 2H), 6.43 (d, / = 3.2 Hz, 1H), 5.15 (s, 2H), 4.39 (t, / = 6.8 Hz, 2H), 3.68 (s, 3H), 2.80 (t, / = 6.8 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 171.58, 155.46, 137.49, 136.35, 128.50, 127.81 , 127.43, 126.96, 123.29, 121.59, 109.98, 101.56, 94.61, 70.77, 51.79, 41.71 , 34.49; MS (CI + ) m/z 309.134 (M + ), 310.142 (MH + ); HRMS calcd. for C19H19NO3 (Μ + ·, Cr/CHi) 309.1365 found 309.1342, for C19H20NO3 (MH + , CI + /CH 4 ) 310.1443 found 310.1416.

Methyl 3-(Indolin-l-yl)propanoate, (3a). Compound 3a synthesised from 2a by Procedure B method I or from indoline 5a by Procedure A, was isolated as a yellow oil in 83 or 76% yield, respectively. J H-NMR (300 MHz, CDCb) ppm δ 7.10-6.99 (m, 2H), 6.617 (t, / = 7.2 Hz, 1H), 6.47 (d, / = 8.1 Hz, 1H), 3.63 (s, 3H), 3.36 (t, / = 6.9 Hz, 2H), 3.28 (t, / = 8.1 Hz, 2H), 2.89 (t, / = 8.1 Hz, 2H), 2.50 (t, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 172.38, 151.63, 129.70, 127.15, 124.25, 117.66, 106.86, 52.85, 51.44, 44.87, 32.05, 28.38.

Methyl 3-(5-Methoxyindolin-l-yl)propanoate, (3b). Compound 3b prepared from 2b by Procedure B method II, was purified by elution through a short silica gel column with EtOAc-hexane (1 : 10->1:7), and was isolated as a yellow oil in 57% yield. ¾-NMR (300 MHz, CDCb) ppm δ 6.89 (d, / = 8.4 Hz, 1H), 6.79-6.72 (m, 2H), 3.77 (s, 3H), 3.70 (s, 3H), 3.55 (t, / = 7.2 Hz, 2H), 3.50 (t, / = 6.9 Hz, 2H), 3.07 (t, / = 7.2 Hz, 2H), 2.73 (t, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 171.76, 156.74, 140.49, 133.76, 113.17, 113.06, 111.66, 55.97, 53.88, 52.09, 48.73, 31.07, 28.70; MS (TOF MS ES + ) m/z 236

Methyl 3-(6-methylindolin-l-yl)propanoate, (3c). Compound 3c synthesised from 2c by Procedure B method III, was isolated as a yellow oil, in approximately quantitative yield. J H-NMR (300 MHz, CD 3 OD) ppm δ 6.88 (d, / = 7.2 Hz, 1H), 6.47 (d, / = 7.2 Hz, 1H), 6.38 (s, 1H), 3.64 (s, 3H), 3.31 (t, / = 6.9 Hz, 2H), 3.22 (t, / = 8.4 Hz, 2H), 2.79 (t, / = 8.1 Hz, 2H), 2.55 (t, / = 6.9 Hz, 2H), 2.22 (s, 3H); 13 C-NMR (75 MHz, CD3OD) ppm δ 174.39, 152.54, 137.93, 128.50, 125.04, 120.45, 109.82, 54.25, 52.14, 46.57, 32.80, 28.86, 21.72.

Methyl 3-(5-Chloroindolin-l-yl)propanoate, (3d). Compound 3d synthesised from 2d by Procedure B method III, was isolated as a red-orange oil in approximately quantitative yield. J H-NMR (300 MHz, CDCb) ppm δ 6.96-6.93 (m, 2H), 6.35 (d, / = 9.0 Hz, 1H), 3.65 (s, 3H), 3.34 (t, / = 6.9 Hz, 2H), 3.31 (t, / = 8.4 Hz, 2H), 2.86 (t, / = 8.4 Hz, 2H), 2.55 (t, / = 7.2 Hz, 2H); 13 C-NMR (75 MHz, CDCI3) ppm δ 172.40, 150.42, 131.69, 126.80, 124.47, 122.00, 107.35, 52.95, 51.59, 44.82, 32.00, 28.19; MS (ES + ) m/z 240 (MH+)

Methyl 3-(6-(Benzyloxy)indolin-l-yl)propanoate, (3f). Compound 3f synthesised from 2f by Procedure B method I, and gave the product as a yellow oil, in 95% yield. J H-NMR (300 MHz, CDCb) ppm δ 7.44-7.23 (m, 5H), 6.90 (d, / = 8.1 Hz, 1H), 6.23 (dd, / = 8.1, 2.1 Hz, 1H), 6.17 (d, / = 2.1 Hz, 1H) 4.98 (s, 2H), 3.65 (s, 3H), 3.39-3.30 (m, 4H), 2.85 (t, / = 8.2 Hz, 2H), 2.56 (t, / = 7.0 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 172.80, 159.32, 153.05, 137.40, 128.46, 127.75, 127.62, 124.43, 122.56, 102.65, 95.56, 70.12, 53.54, 51.74, 44.74, 32.10, 27.77; MS (ΕΓ) m/z 311.152 (M + ), 312.155 (MH + ); HRMS calcd. for C 1 9H2 1 NO3 (Μ + ·, ΕΓ) 311.1521 found 311.1524.

Methyl 3-(Indolin-l-yl)propanoate Hydrochloride, (4a AN1283). The hydrochloride salt 4a was prepared by addition of a solution of HCl (gas)/ether to 3a and was isolated as an orange hygroscopic solid in approximately quantitative yield. ¾-NMR (300 MHz, CD3OD) ppm δ 7.63-7.60 (m, 1H), 7.54-7.47 (m, 3H), 4.00 (t, / = 7.5 Hz, 2H), 3.86 (t, / = 6.9 Hz, 2H), 3.74 (s, 3H), 3.37 (t, / = 7.5 Hz, 2H), 2.97 (t, / = 6.9 Hz, 2H; 13 C-NMR (75 MHz, CD 3 OD) ppm δ 172.10, 141.41 , 136.65, 131.77, 129.94, 127.68, 119.88, 55.54, 53.33, 52.84, 30.25, 28.92.

Methyl 3-(5-Methoxyindolin-l-yl)propanoate Hydrochloride, (4b AN1297).

Compound 4b was prepared by addition of HCl (g) to 3b (0.11 g/ 0.47mmol) in dry diethyl ether (12 mL). The precipitate was isolated as a dark red oil in approximately quantitative yield. 1 H-NMR (300 MHz, CD 3 OD) ppm δ 7.52 (d, / = 8.7 Hz, 1H, 7.06- 6.98 (m, 2H), 4.02 (t, / = 7.5 Hz, 2H), 3.84 (s, 3H), 3.81 (t, / = 7.2 Hz, 2H), 3.73 (s, 3H), 3.34 (t, / = 7.5 Hz, 2H), 2.97 (t, / = 7.2 Hz, 2H); 13 C-NMR (75 MHz, CD3OD) ppm δ 172.01 , 163.20, 138.55, 133.45, 121.00, 115.89, 112.13, 56.48, 56.05, 53.64, 52.87, 30.20, 29.11 ; MS (TOF MS ES + ) m/z 236.

Methyl 3-(6-Methylindolin-l-yl)propanoate Hydrochloride, (4c AN1296). The hydrochloride of 4c obtained by addition of a solution of HCl (gas)/ether to 3c was isolated as a yellow oil in approximately quantitative yield. ¾-NMR (300 MHz, CD3OD) ppm δ 7.46 (s, 1H), 7.38-7.30 (m, 2H), 4.00 (t, / = 7.5 Hz, 2H), 3.84 (t, / = 7.2 Hz, 2H), 3.70 (s, 3H), 3.30 (t, / = 7.5 Hz, 2H), 3.00 (t, / = 6.9 Hz, 2H), 2.40 (s, 3H); 13 C-NMR (75 MHz, CD3OD) ppm δ 171.75, 140.93, 140.55, 133.50, 132.68, 127.12, 120.34, 55.53, 53.19, 52.79, 30.16, 28.52, 21.34.

Methyl 3-(5-Chloroindolin-l-yl)propanoate p-Toluenesulfonate, (4d AN1285).

Compound 4d was prepared by addition of p-TSA to a solution of 3d in i-butyl methyl ether. The ethereal solution was evaporated to give 4d as a brown oil that was found that the salt contained a -50% excess of p-TSA, detected by NMR. ¾-NMR (300 MHz, CD3OD) ppm δ 7.62 (d, / = 8.4 Hz, 4H), 7.53 (d, / = 8.4 Hz, 1H), 7.35-7.30 (m, 2H), 3.98 (t, / = 7.5 Hz, 2H), 3.79 (t, / = 7.2 Hz, 2H), 3.65 (s, 3H), 3.24 (t, / = 7.5 Hz, 2H), 2.91 (t, /= 7.2 Hz, 2H), 2.29 (s, 6H); 13 C-NMR (75 MHz, CD3OD) ppm δ 171.53, 142.60, 141.73, 139.59, 138.86, 137.05, 129.76, 129.62, 127.39, 126.60, 121.47, 55.77, 53.27, 52.72, 30.03, 28.65, 27.14, 21.32; MS (ES + ) m/z 240 (MH+).

Methyl 3-(6-(Benzyloxy)indolin-l-yl)propanoate p-Toluenesulfonate, (4f AN1279).

Compound 4f was prepared by addition of p-TSA (0.13 g, 0.7 mmol) to 3f (0.21 g, 0.7 mmol) in i-butyl methyl ether (10 mL) to give the product as a hygroscopic white solid, having 2 equivalents of p-TSA. It should be noted that an H— >D exchange took place when 4f was dissolved in CD3OD in an NMR tube. The exchange was detected by NMR already after 5 min, and was shown to take place at position 5 to give 4f-A, and partly at both positions 5 and 7 to give 4f-B. J H-NMR (600 MHz, CD3OD) ppm δ 7.66 (d, / = 8.4 Hz, 4.5H), 7.43 (d, / = 7.2 Hz, 2H), 7.36 (t, / = 7.2 Hz, 2H), 7.33 (s, 1H), 7.32-7.29 (m, 1H), 7.21 (d, / = 8.4 Hz, 4.5H), 7.15 (s, 0.4H), 5.10 (s, 2H), 3.93 (t, / = 8.2 Hz, 2H), 3.77 (t, / = 8.2 Hz, 2H), 3.71 (s, 3H), 3.20 (t, / = 8.2 Hz, 2H), 2.86 (t, / = 8.2 Hz, 2H), 2.35 (s, 7H); 13 C-NMR (150 MHz, CD 3 OD) ppm δ 172.30, 160.69, 143.50, 143.01, 141.75, 138.00, 129.84, 129.62, 129.15, 128.76, 127.88, 127.82, 126.97, 105.67, 71.70, 56.26, 52.91, 52.82, 30.40, 28.28, 21.33; MS (for SZ-I-107) (EI + ) m/z 311.152 (M + ), 312.155 (MH + ); HRMS calcd. for C 1 9H2 1 NO3 (Μ + ·, ΕΓ) 311.1521 found 311.1524 and calcd for CwHaiNCfeNa (MNa + , ESI + ) 334.1419 found 334.1422.

3-(Indolin-l-yl)propan-l-ol, (AN1400 free base). (Yushi et al., 2015) Compound 6a free base prepared from 3a by Procedure F, was isolated as a dark oil in 98% yield. J H- NMR (300 MHz, CDCb) ppm δ 7.04-6.99 (m, 2H), 6.63 (t, J = 7.5 Hz, 1H), 6.49 (d, J = 7.5 Hz, 1H), 4.04 (bs, 1H), 3.66 (t, J = 6.0 Hz, 2H), 3.25 (t, J = 8.1 Hz, 2H), 3.10 (t, J = 6.3 Hz, 2H), 2.88 (t, J = 8.1 Hz, 2H), 1.78 (quint, J = 6.3 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 152.33, 129.94, 127.07, 124.20, 117.83, 107.35, 60.63, 53.24, 46.88, 29.78, 28.35; MS (ES + ) m/z 178.2 (MH+).

3-(5-Methoxyindolin-l-yl)propan-l-ol, (6b free base). Compound 6b free base prepared from 3b by Procedure F was isolated as a yellow oil in 85% yield. J H-NMR (300 MHz, CDCb) ppm δ 6.72 (s, 1H), 6.63-6.59 (m, 1H), 6.48-6.45 (m, 1H), 3.74 (t, / = 6.0 Hz, 2H), 3.70 (s, 3H), 3.22 (t, / = 8.1 Hz, 2H), 3.06 (t, / = 6.6 Hz, 2H), 2.87 (t, / = 7.8 Hz, 2H), 1.79 (quint, / = 6.6 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 153.14, 146.69, 131.78, 111.70, 111.70, 108.34, 61.49, 55.79, 54.23, 48.97, 29.82, 28.76; MS (ES + ) m/z 208 (MH+); HRMS calcd. for C12H17NO2 (MH + , ESI + ) 208.1332 found 208.1333.

3-(Indolin-l-yl)propan-l-ol Hydrochloride, (6a AN1400). Compound 6a was prepared by addition of a solution of HCl (gas)/ether to 6a free base and was isolated as a greyish hygroscopic solid in approximately quantitative yield. J H-NMR (600 MHz, CD3OD) ppm δ 7.61 (d, / = 7.8 Hz, 1H), 7.53-7.52 (m, 2H), 7.51-7.48 (m, 1H), 4.01 (bt, 2H), 3.74 (t, / = 6.0 Hz, 2H), 3.67 (bt, 2H), 3.37 (t, / = 7.8 Hz, 2H), 2.02 (quint, / = 7.2 Hz, 2H); 13 C- NMR (150 MHz, CD3OD) ppm δ 141.66, 136.68, 131.79, 129.96, 127.71, 119.97, 60.00, 56.17, 55.50, 29.00, 28.48; MS (ES + ) m/z 178.2 (MH+).

l\^N HCl

^~^-OH

3-(5-Methoxyindolin-l-yl)propan-l-ol Hydrochloride, (6b AN1299 hydrochloride).

Compound 6b was prepared by addition of a solution of HCl (gas)/ether to 6b free base and was isolated as a brown hygroscopic solid in approximately quantitative yield. ¾- NMR (700 MHz, CD3OD, at 275 °K) ppm δ 7.53 (d, / = 9.1 Hz, 1H), 7.05 (d, / = 2.1 Hz, 1H), 6.97 (dd, / = 9.1, 2.8 Hz, 1H), 4.16 (br, 1H), 3.9-3.8 (br, 2H), 3.82 (s, 3H), 3.71 (t, / = 6.3 Hz, 2H), 3.45 (br, 1H), 3.36 (br, 1H), 3.27 (br, 1H), 2.07 (br, 1H), 1.98 (br, 1H); 13 C-NMR (176 MHz, CD3OD, 275 °K) ppm δ 162.66, 138.35, 133.53, 120.92, 115.542, 111.80, 59.58, 56.37, 55.73, 55.45, 29.03, 28.33; MS (ES + ) m/z 208 (MH+); HRMS calcd. for C12H17NO2 (MH + , ESI + ) 208.1332 found 208.1333.

3-(lH-Indol-l-yl)propanenitrile, (7a). (Wittig et al., 1958), Compound 7a was synthesised from indole by Procedure C, and was isolated as yellow oil in approximately quantitative yield. J H-NMR (300 MHz, CDCb) ppm δ 7.59 (dd, / = 7.8, 0.9 Hz, 1H) 7.20- 7.13 (m, 2H), 7.13-7.07 (m, 1H), 6.98 (d, / = 3.0 Hz, 1H), 6.47 (d, / = 3.0 Hz, 1H), 4.11 (t, / = 6.6 Hz, 2H), 2.49 (t, / = 6.6 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 135.18, 128.67, 127.47, 121.91, 121.51, 119.88, 117.45, 108.75, 102.35, 41.58, 118.72; MS (ΕΓ) m/z 170.085 (M + ), 171.084 (MH + ); HRMS calcd. for CnHioN 2 (M + -, ΕΓ) 170.0844 found 170.0853.

3-(7-(Benzyloxy)-lH-indol-l-yl)propanenitrile, (7g). Compound 7g prepared from 7- benzyloxyindole by Procedure C, was purified by crystallisation from DCM-ether, and was isolated as a bright orange solid, mp 69-71 °C, in approximately quantitative yield. ¾-NMR (400 MHz, CDCb) ppm δ 7.47-7.35 (m, 5H), 7.24 (dd, / = 8.0, 0.8 Hz, 1H), 7.02 (t, / = 8.0 Hz, 1H), 7.01 (d, / = 3.2 Hz, 1H), 6.75 (d, / = 8.0 Hz, 1H), 6.47 (d, / = 3.2 Hz, 1H), 5.17 (s, 2H), 4.56 (t, / = 6.8 Hz, 2H), 2.70 (t, / = 6.8 Hz, 2H); 13 C-NMR (100 MHz, CDCb) ppm δ 146.34, 136.60, 131.69, 129.14, 128.97, 128.55, 129.34, 125.00, 120.68, 117.59, 114.49, 103.82, 102.66, 70.69, 45.28, 20.98; MS (EI + ) m/z 276.123 (M + ), 277.133 (MH + ), 185.059 (M-Bn); HRMS calcd. for Ci 8 Hi 6 N 2 0 (Μ + ·, EI + ) 276.1263 found 276.1226.

3-(Indolin-l-yl)propanenitrile, (8a). (Astil and Boekelheide 1958). Compound 8a synthesised from 7a using Procedure B Method I, was isolated as a yellow oil in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.12-7.07 (m, 2H), 6.72 (t, / = 7.5 Hz, 1H), 6.48 (d, / = 7.8 Hz, 1H), 3.46-3.39 (m, 4H), 3.00 (t, / = 8.4 Hz, 2H), 2.59 (t, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 150.76, 129.74, 128.54, 124.64, 118.64, 118.34, 106.67, 52.90, 45.17, 28.43, 11.01 ; MS (EI + ) m/z 172.106 (M + ), 173.109 (MH + ); HRMS calcd. for C11H12N2 (Μ + ·, ΕΓ) 172.1000 found 172.1061.

3-(7-(Benzyloxy)indolin-l-yl)propanenitrile, (8g). Compound 8g synthesised from 7g by Procedure B Method I, was isolated as a yellow oil in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.46-7.26 (m, 5H), 6.77-6.63 (m, 3H), 4.98 (s, 2H), 3.63 (t, / = 6.9 Hz, 2H), 3.40 (t, / = 8.7 Hz, 2H), 2.94 (t, / = 8.7 Hz, 2H), 2.42 (t, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 144.98, 138.69, 136.69, 132.04, 128.58, 128.03, 127.53, 119.83, 118.99, 118.01 , 112.11 , 70.63,54.08, 47.23, 29.06, 16.67; MS (EI + ) m/z 27S. U6 (M + ), 279.156 (MH + ); HRMS calcd. for Ci 8 Hi 8 N 2 0(M + -, EI + ) 278.1419 found 278.1462.

3-(Indolin-l-yl)propan-l-amine, (9a). (a) Petrovna et al., 2010; b) Shapiro et al., 1959). Compound 9a synthesised from 8a by Procedure E, was isolated as a yellow oil in 68% yield. J H-NMR (300 MHz, CDCb) ppm δ 6.90-6.86 (m, 2H), 6.48 (t, / = 7.2 Hz, 1H), 6.29 (t, / = 7.2 Hz, 1H), 3.09 (t, / = 8.1 Hz, 1H), 2.87 (t, / = 6.9 Hz, 1H), 2.74 (t, / = 8.1 Hz, 2H), 2.50 (t, / = 6.9 Hz, 2H), 1.47 (t, / = 7.2 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 152.02, 129.84, 126.75, 124.02, 116.99, 106.43, 52.63, 48.44, 39.43, 30.42, 28.03; MS (ΕΓ) m/z 176.134 (M + ); HRMS calcd. for CnHi 6 N 2 + ·, ΕΓ) 176.1313 found 176.1337.

3-(7-(Benzyloxy)indolin-l-yl)propan-l-amine, (9g). Compound 9g was prepared from 8g by Procedure E, and was isolated as a yellow oil in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.50-7.17 (m, 5H), 6.83-6.60 (m, 3H), 4.99 (s, 2H), 3.41 (t, / = 6.9 Hz, 2H), 3.33 (t, / = 8.4 Hz, 2H), 2.95 (t, / = 8.4 Hz, 2H), 2.61 (t, / = 6.9 Hz, 2H), 1.87 (bs, 2H), 1.61 (quint, / = 6.9 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 145.44, 140.73, 137.17, 132.35, 128.55, 128.04, 127.79, 119.25, 118.02, 112.42, 70.82, 53.91, 49.18, 39.79, 31.54, 29.23; MS (ESI + ) m/z 283 (M + ); HRMS calcd. for Ci8H 2 2N 2 ONa (M + -, ESI + ) 305.1630 found 305.1626.

3-(Indolin-l-yl)propanenitrile di-p-Toluenesulfonate, (10a AN1264). Compound 10a was prepared by addition of /?-TSA (4.3 g, 22 mmol) to 9a (2 g, 11 mmol) in i-butyl methyl ether (60 mL). The precipitate was crystallized from DCM-ether and was isolated as a hygroscopic white solid, which contained an approximate 20% excess of /?-TSA detected by NMR. J H-NMR (300 MHz, CD 3 OD) ppm δ 7.66 (d, / = 8.1 Hz, 4H), 7.58 (bd, / = 7.8 Hz, 1H), 7.54-7.37 (m, 3H), 7.2 (d, / = 8.1 Hz, 4H), 3.97 (t, / = 7.8 Hz, 2H), 3.65 (".", /= 8.1 Hz, 2H), 3.29 (m, 2H), 3.09 (t, / = 7.5 Hz, 2H), 2.34 (s, 6H), 3.09 (quint, / = 8.1 Hz, 2H); 13 C-NMR (75 MHz, CD3OD) ppm δ 143.14, 141.98, 140.91, 136.63, 131.97, 129.96, 127.68, 126.90, 120.17, 55.40, 54.60, 37.80, 28.97, 24.01, 21.34; MS (ΕΓ) m/z 176.137 (M + ), 177.142 (MH + ); HRMS calcd. for CnHi 6 N 2 (M + -, ΕΓ) 176.1313 found 176.1370.

3-(7-(Benzyloxy)indolin-l-yl)propan-l-amine di-p-Toluenesulfonate, (lOg AN1276).

Compound lOg was prepared by addition of /?-TSA to a solution of 9g in i-butyl methyl ether. The ethereal solution was evaporated and lOg was crystallized from DCM-ether as a hygroscopic white solid in approximately quantitative yield. J H-NMR (300 MHz, CD3OD) ppm δ 7.69-7.65 (m, 4H), 7.52-7.33 (m, 6H), 7.20-7.14 (m, 5H), 7.03 (dd, / = 7.5, 0.6 Hz, 1H), 4.01 (t, / = 7.5 Hz, 2H), 3.60 ("t", / = 7.2 Hz, 2H), 3.35-3.30 (m, 2H), 2.96 (t, / = 7.5 Hz, 2H), 2.35 (s, 6H), 2.12-2.06 (m, 2H); 13 C-NMR (75 MHz, CD3OD) ppm δ 151.32, 143.28, 141.85, 138.83, 137.13, 133.91, 129.90, 129.82, 129.55, 129.08, 127.98, 126.86, 119.13, 113.67, 72.12, 66.84, 55.44, 54.01, 37.80, 29.32, 23.78, 21.31, 15.44.

3-(Indolin-l-yl)-N-isopropylpropan-l-amine, (11a). Compound 11a prepared from 16a by Procedure B method III, was isolated as a brown oil, in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.05-7.00 (m, 2H), 6.64-6.59 (m, 1H), 6.46- 6.43 (m, 1H), 3.29 (t, / = 8.4 Hz, 2H), 3.07 (t, / = 7.2 Hz, 2H), 2.91 (t, / = 8.4 Hz, 2H), 2.77 (septet, / = 6.3 Hz, 1H), 1.74 (quint, / = 7.2 Hz, 2H), 1.04 (d, / = 6.3 Hz, 6H); 13 C- NMR (75 MHz, CDCb) ppm δ 152.56, 129.88, 127.17, 124.28, 117.40, 106.87, 53.11, 48.70, 47.59, 45.38, 28.46, 28.10, 22.79.

N-Isopropyl-3-(5-methoxyindolin-l-yl)propan-l-amine, (lib). Compound lib synthesised from 16b by Procedure B method I, was isolated as a yellow oil in 75% yield. ¾-NMR (300 MHz, CDCb) ppm δ J H-NMR (300 MHz, CDCb) ppm δ 6.72 (s, 1H), 6.63-6.60 (m, 1H), 6.43-6.40 (m, 1H), 3.71 (s, 3H), 3.25 (t, / = 8.1, 2H), 3.02 (t, / = 6.9, 2H), 2.89 (t, / = 8.1, 2H), 2.82 (septet, / = 6.3 Hz, 1H), 2.72 (t, / = 6.9, 2H), 2.512-2.493 (m, 2H), 1.78 (quint, / = 7.2 Hz, 2H), 1.06 (d, / = 6.3 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 152.76, 146.87, 131.51, 111.79, 111.64, 107.57, 55.81, 53.89, 48.95, 48.77, 45.41, 28.75, 27.91, 22.56; MS (ES + ) m/z 249.2 (MH+). l-(3-(Isopropylamino)propyl)indolin-7-ol, (llg). To a methanolic solution of a 9g (1 eq), was added 10% Pd/C. The mixture was stirred under ¾ at 4 atm. (Rubino et al., 2011), pressure for 23 h at room temperature in the presence of traces of acetone in the Parr hydrogenator. The mixture was then filtered through celite and concentrated to give comound llg as a brown oil, in -90% yield, and was used without further purification.

¾-NMR (300 MHz, CDC1 3 ) ppm δ 6.70-6.56 (m, 3H), 3.36-3.30 (m, 4H), 2.98 (t, / = 8.4 Hz, 2H), 2.85 (t, / = 5.7 Hz, 2H), 2.83 (septet, / = 6.6 Hz, 1H), 1.67 (quint, / = 5.7 Hz, 2H), 1.10 (d. /= 6.6 Hz, 6H); 13 C-NMR (100 MHz, CDCl 3 )ppm 5 144.11, 141.06, 131.46, 120.01, 117.38, 115.74, 54.53, 49.04, 48.80, 42.86, 29.61, 27.59, 21.77.

3-(Indolin-l-yl)-N-isopropylpropan-l-amine di-p-Toluenesulfonate (12a AN1284).

Compound 12a was prepared by addition of p-TSA to a solution of 11a in i-butyl methyl ether. The ethereal solution was evaporated to give 18a as an hygroscopic brown solid, in 23% yield. J H-NMR (300 MHz, CD3OD) ppm δ 7.78 (d, / = 8.1 Hz, 5H), 7.69-7.66 (m, 1H), 7.59-7.51 (m, 3H), 7.31 (d, / = 7.8 Hz, 5H), 4.06 (t, / = 7.5 Hz, 2H), 3.76 ("t", / = 8.1 Hz, 2H), 3.47-3.36 (m, 3H), 3.26 (t, / = 7.8 Hz, 2H), 2.45 (s, 8H), 2.33 (quint, / = 7.8 Hz, 2H) 1.40 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CD3OD) ppm δ 143.30, 141.86, 141.14, 136.47, 131.64, 129.85, 127.57, 126.85, 119.93, 55.30, 54.42, 52.28, 42.96, 28.93, 27.19, 23.14, 21.31, 19.19.

N-Isopropyl-3-(5-methoxyindolin-l-yl)propan-l-amine di-Hydrochloride, (12b AN1298). Compound 12b obtained by addition of a solution of HCl (gas)/ether to lib, was isolated as a hygroscopic yellow solid in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.08-7.05 (m, 1H), 6.91-6.90 (m, 1H), 6.85-6.82 (m, 1H), 3.77 (s, 3H), 3.67 (t, / = 7.8, 2H), 3.42-3.34 (m, 3H), 3.19-3.11 (m, 4H), 2.12 (quint, / = 7.5 Hz, 2H), 1.35 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 159.54, 139.55, 136.10, 116.08, 114.58, 112.34, 56.41, 55.43, 52.41, 52.08, 43.53, 29.41, 24.07, 19.25; MS (ES + ) m/z 249.2 (MH+).

l-(3-(Isopropylamino)propyl)indolin-7-ol di-p-Toluenesulfonate, (12g AN1280).

Compound 12g was prepared by addition of p-TSA (0.39 g, 2 mmol) to l lg (0.2 g, 1 mmol) in i-butyl methyl ether (10 mL). The precipitate was crystallised from DCM-ether as a hygroscopic pink solid, and was isolated in quantitative yield. J H-NMR (700 MHz, CD3OD+D2O) ppm δ 7.69 (d, / = 8.4 Hz, 4H, H-20), 7.32 (t, / = 7.7 Hz, 1H), 7.25 (d, / = 8.4 Hz, 4H), 6.94 (dd, / = 7.7, 0.7 Hz, 1H), 6.91 (d, / = 7.7 Hz, 1H), 3.95 (t, / = 7.7 Hz, 2H), 3.66 ("t", / = 7.7 Hz, 2H), 3.37 (septet, / = 6.3 Hz, 1H), 3.34 (t, / = 7.7 Hz, 2H), 3.13 (t, / = 7.7 Hz, 2H), 2.37 (s, 6H), 2.16 (quin, / = 7.7 Hz, 2H), 1.33 (d, / = 6.3 Hz, 6H); 13 C-NMR (176 MHz, CD3OD+D2O) ppm δ 149.98, 142.95, 141.99, 138.18, 132.90, 129.92, 127.00, 126.82, 117.74, 116.41, 55.22, 53.35, 52.19, 43.02, 29.31 , 23.06, 21.34, 19.20.

3-(lH-indol-l-yl)-N-isopropylpropanamide, (13a). Compound 13a synthesised from indole by Procedure D, was isolated as an orange oil in 59% yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.61 (d, / = 7.8 Hz, 1H), 7.36-7.33 (m, 1H), 7.20 (td, / = 7.8, 0.9 Hz, 1H), 7.13-7.06 (m, 2H), 6.46 (d, / = 2.7 Hz, 1H), 5.06 (bs, 1H), 4.46 (t, / = 6.6 Hz, 2H), 3.95 (septet, / = 6.6 Hz, 1H), 2.54 (t, / = 6.6 Hz, 2H), 0.96 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 169.44, 135.66, 128.83, 128.28, 121.66, 121.15, 119.55, 109.30, 101.56, 42.71, 41.59, 37.72, 22.53; MS (ES + ) m/z 253 (MNa) +

N-Isopropyl-3-(5-methoxy-lH-indol-l-yl)propanamide, (13b). Compound 13b synthesised from 5-methoxyindoline by Procedure D, was isolated as an orange solid in 85% yield, mp 68-71 °C. ¾-NMR (300 MHz, CDCb) ppm δ 7.17 (d, / = 8.7 Hz, 1H), 7.04-7.00 (m, 2H), 6.82 (dd, / = 8.7, 2.4 Hz, 1H), 6.35 (d, / = 3 Hz, 1H), 5.73-5.70 (m, 1H), 4.33 (t, / = 6.3 Hz, 2H), 3.89 (septet, / = 6.6 Hz, 1H), 3.62 (s, 3H), 2.49 (t, / = 6.3 Hz, 2H), 0.93 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 170.10, 153.95, 130.88, 129.01 , 128.55, 111.79, 109.92, 102.69, 101.04, 55.75, 42.68, 41.60, 37.43, 22.16; MS (ES + ) m/z 261 (MH+), 283 (MNa) + , 299 (MK) +

3-(5-Chloro-lH-indol-l-yl)-N-isopropylpropanamide, (13d). Compound 13d synthesised from 5-chloroindole by Procedure D, was isolated bright orange solid, in approximately quantitative yield, mp 122-125 °C. J H-NMR (300 MHz, CDCb) ppm δ 7.51 (s, 1H), 7.22-7.18 (m, 1H), 7.10-7.05 (m, 2H), 6.35-6.34 (m, 1H), 4.33 (t, / = 6.6 Hz, 2H), 3.90 (septet, / = 6.6 Hz, 1H), 2.46 (t, / = 6.6 Hz, 2H), 0.94 (d. / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 169.00, 133.91 , 129.49, 129.36, 124.88, 121.53, 120.08, 110.20, 100.89, 42.53, 41.09, 37.06, 22.19; MS (ES + ) m/z 265 (MH+), 287 (MNa) +

3-(6-Fluoro-lH-indol-l-yl)-N-isopropylpropanamide, (13e). Compound 13e synthesised from 6-fluoroindole by Procedure D, was isolated as a white solid, mp 86-87 °C, in approximately quantitative yield. ¾-NMR (600 MHz, CDCb) ppm δ 7.50 (dd, / = 9.0, 5.4 Hz, 1H), 7.08 (d, / = 3.0 Hz, 1H), 7.03 (dd, / = 9.6, 1.8 Hz, 1H), 6.86 (td, / = 9.6, 2.4 Hz, 1H), 6.43 (d, / = 3.0 Hz, 1H), 5.20-5.09 (m, 1H), 4.42 (t, / = 6.6 Hz, 2H), 3.97 (septet, / = 6.6 Hz, 1H), 3.69 (s, 3H), 2.55 (t, / = 6.6 Hz, 2H), 0.98 (d, / = 6.6 Hz, 6H); 13 C-NMR (150 MHz, CDCb) ppm δ 169.20, 160.54+158.96, 135.65+135.57, 128.64+128.61 , 125.10, 121.75+121.69, 108.28+108.11, 101.66, 95.78+95.61, 42.75, 41.60, 37.31, 22.42.

3-(Indolin-l-yl)-N-isopropylpropanamide, (14a). Compound 14a synthesised from 13a by Procedure B method III, was isolated as a yellow oil in 79% yield. J H-NMR (400 MHz, CDCb) ppm δ 7.06-7.00 (m, 2H), 6.71-6.65 (m, 2H) 6.50 (d, / = 7.6 Hz, 1H), 3.99 (septet, / = 6.6 Hz, 1H), 3.33-3.20 (m, 4H), 2.90 (t, / = 8.0 Hz, 2H), 2.41 (t, / = 6.4 Hz, 2H), 1.08 (d, / = 6.8 Hz, 6H); 13 C-NMR (100 MHz, CDCb) ppm δ 171.32, 151.47, 130.11 , 127.21, 124.47, 118.48, 107.47, 53.09, 46.15, 41.22, 34.01, 28.38, 22.19; MS (ES + ) m/z 233 (MH+), 255 (MNa) +

3-(5-Chloro-lH-indol-l-yl)-N-isopropylpropanamide, (14d). Compound 14d synthesised from 13d by Procedure B method III, was isolated as an orange oil, in 93% yield. J H-NMR (300 MHz, CDCb) ppm δ 6.97-6.93 (m, 2H), 6.50-6.47 (m, 1H), 6.36 (d, / = 8.1 Hz, 1H), 4.00 (septet, / = 6.6 Hz, 1H), 3.32 (t, / = 7.5 Hz, 4H), 2.88 (t, / = 8.1 Hz, 2H), 2.40 (t, / = 6.9 Hz, 2H), 1.10 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 170.76, 150.48, 131.82, 126.80, 124.53, 122.21 , 107.54, 53.14, 45.69, 41.22, 34.08, 28.22, 22.39; MS (ES + ) nt/z 267 (MH+), 289 (MNa) +

3-(6-Fluoroindolin-l-yl)-N-isopropylpropanamide, (14e). Compound 14e synthesised from 13e by Procedure B method III, was isolated as a white solid after sublimation, in 5% yield, mp 69-70 °C. ¾-NMR (300 MHz, Acetone-d) ppm δ 6.93-6.90 (m, 1H), 6.25- 6.18 (m, 2H), 3.96 (septet, / = 6.6 Hz, 1H), 3.43 (t, / = 8.4 Hz, 2H), 3.38 (t, / = 6.9 Hz, 2H), 2.85 (t, / = 8.4 Hz, 2H), 2.38 (t, / = 6.9 Hz, 2H), 1.08 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, Acetone-d6) ppm δ 170.56, 165.98, 162.83, 154.93+154.91 , 126.13, 125.33+125.19, 103.09+102.79, 95.34+94.97, 54.07, 41.62+41.51 , 34.49+34.45, 28.29, 22.79; MS (ES + ) nt/z 251 (MH+), 273 (MNa) +

3-(Indolin-l-yl)-N-isopropylpropanamide Hydrochloride, (15a AN1292). Compound 15a was prepared by addition of 3N HCI to 14a in ethyl acetate and was evaporated. The precipitate was crystalized and was isolated as a white-brown solid, mp 92-93 °C, in approximately quantitative yield. ¾-NMR (300 MHz, CD 3 OD) ppm δ 7.64 (d, / = 7.2 Hz, 1H), 7.53-7.46 (m, 3H), 4.06 (t, / = 7.2 Hz, 2H), 3.89 (septet, / = 6.6 Hz, 1H), 3.87 (t, / = 6.9 Hz, 2H), 3.39 (t, / = 6.9 Hz, 2H), 2.87 (t, / = 6.6 Hz, 2H), 1.14 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CD3OD) ppm δ 169.96, 140.59, 136.64, 131.70, 129.66, 127.47, 120.18, 54.96, 53.65, 42.72, 31.22, 28.91 , 22.44; MS (ES + ) m/z 233 (MH+), 255 (MNa) +

3-(5-Chloro-lH-indol-l-yl)-N-isopropylpropanamide Hydrochloride, (15d AN1287). Compound 15d was prepared by addition of 3N HCl to 14d in EtOAc and was evaporated. The precipitate was isolated as a dark yellow oil in approximately quantitative yield. J H-NMR (300 MHz, CD3OD) ppm δ 7.66-7.63 (m, 1H), 7.56-7.50 (m, 2H), 4.08 (t, / = 7.5 Hz, 2H), 3.90 (septet, / = 6.6 Hz, 1H), 3.86 (t, / = 6.9 Hz, 2H), 3.41 (t, / = 7.5 Hz, 2H), 2.83 (t, / = 6.9 Hz, 2H), 1.14 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CD3OD) ppm δ 170.14, 139.73, 139.25, 137.35, 129.91, 127.685, 121.73, 55.57, 53.94, 42.82, 31.22, 29.03, 22.50; MS (ES + ) m/z 267 (MH+), 289 (MNa) +

3-(6-Fluoroindolin-l-yl)-N-isopropylpropanamide Hydrochloride, (15e AN1294).

Compound 15e obtained by addition of a solution of HCl (gas)/ether to 14e was isolated as a hygroscopic white-pink solid, in approximately quantitative yield. ¾-NMR (400 MHz, CD3OD) ppm δ 7.52 (dd, / = 8.4, 5.4 Hz, 1H), 7.46 (dd, / = 8.4, 2.0 Hz, 1H), 7.26 (td, / = 8.8, 2.4 Hz, 1H), 4.06 (t, / = 7.6 Hz, 2H), 3.98 (septet, / = 6.4 Hz, 1H), 3.84 (t, / = 6.8 Hz, 2H), 3.33 (t, / = 7.6 Hz, 2H), 2.78 (t, / = 6.8 Hz, 2H), 1.15 (d, / = 6.8 Hz, 6H); 13 C-NMR (100 MHz, CD3OD) ppm δ 170.48, 165.03+162.58, 142.71 , 132.56, 128.89+128.80, 118.66+118.44, 108.09+107.81 , 56.24, 53.90, 42.85, 31.17, 28.49, 22.54; MS (ES + ) nt/z 251 (MH+), 273 (MNa) +

3-(lH-Indol-l-yl)-N-isopropylpropan-l-amine, (16a). Compound 16a prepared from 13a by Procedure F, was isolated as a brown oil in 50% yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.61-7.59 (m, 1H), 7.33-7.31 (m, 1H), 7.19-7.14 (m, 1H), 7.09-7.05 (m, 2H), 6.46 (d, / = 3.3 Hz, 1H), 4.13 (t, / = 6.9 Hz, 2H), 2.68 (septet, / = 6.3 Hz, 1H), 2.52 (t, / = 6.9 Hz, 2H), 1.93 (quint, / = 6.9 Hz, 2H), 0.99 (d, / = 6.3 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 135.86, 128.50, 127.76, 121.29, 120.83, 119.16, 109.32, 100.97, 48.77, 44.35, 44.11, 41.05, 30.61, 22.65. MS (ES + ) m/z 217.2 (MH+), 253 (MNa) +

N-Isopropyl-3-(5-methoxy-lH-indol-l-yl)propan-l-amine, (16b). Compound 16b synthesised from 13b by Procedure F, was isolated as a yellow oil in 50% yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.17 (d, / = 9 Hz, 1H), 7.00 (d, / = 15.3 Hz, 1H), 6.82 (dd, / = 9, 2.1 Hz, 1H), 6.34 (d, 2.4 Hz, 1H), 4.04 (t, / = 6.9 Hz, 2H), 3.75 (s, 3H), 2.62 (septet, / = 6.2 Hz, 1H), 1.83 (quint, / = 6.7 Hz, 2H), 0.96 (d, / = 6.6 Hz, 6H); 13 C-NMR (75 MHz, CDCb) ppm δ 155.50, 130.91 , 128.49, 127.90, 111.23, 109.72, 101.98, 100.08, 55.14, 48.27, 44.01, 43.79, 30.55, 22.56; MS (ES + ) m/z 247.1 (MH+).

3-(lH-Indol-3-yl)propanamide, (17). To a solution of 3-indole propionic acid (IP A) (1 g, 4.93 mmol) in THF (15 mL) was added carbonyldiimidazole (1.1 g, 5.9 mmol) at room temperature. After stirring for 45 min, a solution of 30% NH40H (15 mL). The resulting reaction mixture was stirred for 16 h and was then evaporated. The residue was dissolved in DCM and washed with water and saturated aqueous NaCl. The organic layer was dried over Na2SC¼ and evaporated. Compound 19 was isolated as a white solid, mp 125-127, in 70% yield. J H-NMR (300 MHz, CDCb) ppm δ 9.99 (bs, 1H), 7.59-7.56 (m, 1H), 7.37- 7.35 (m, 1H), 7.13-6.68 (m, 3H), 6.79 (bs, 1H), 6.17(bs, 1H), 3.04 (t, / = 7.5 Hz, 2H), 2.87 (bs, 1H), 2.56 (t, / = 7.5 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 175.00, 137.74, 128.43, 122.81 , 122.01 , 119.29, 115.61 , 112.00, 37.06, 21.88.

3-(lH-Indol-3-yl)propan-l-amine, (18). Compound 18 was synthesised from 17 by Procedure F, to give 18 as a yellow oil, in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 9.35 (bs, 1H), 7.52-7.50 (m, 1H), 7.22-7.19 (m, 1H), 7.12-7.01 (m, 2H), 6.74 (s, 1H), 2.77 (bs, 2H), 2.64 (t, / = 7.2 Hz, 2H), 2.56 (t, / = 7.2 Hz, 2H), 1.69 (quint, / = 7.2 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 136.27, 127.11, 121.43, 121.16, 118.40, 118.38, 114.86, 111.08, 61.45, 41.40, 33.36, 29.61, 22.14, 13.86.

3-(Indolin-3-yl)propan-l-amine, (19). Compound 19 was synthesised from 18 by Procedure B Method II, and was purified by elution through a short silica gel column with EtOAc-hexane (1 :4), and was isolated as a yellow oil in 6% yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.05-7.03 (m, 2H), 6.70 (t, / = 7.5 Hz, 1H), 6.60 (d, / = 7.8Hz, 1H), 6.74 (s, 1H), 2.77 (bs, 2H), 2.64 (t, / = 7.2 Hz, 2H), 2.56 (t, / = 7.2 Hz, 2H), 1.69 (quint, / = 7.2 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 136.27, 127.11, 121.43, 121.16, 118.40, 118.38, 114.86, 111.08, 61.45, 41.40, 33.36, 29.61, 22.14, 13.86.

3-(Indolin-3-yl)propan-l-amine di-p-Toluenesulfonate, (20 AN1282). Compound 20 was prepared by addition of p-TSA to a solution of 19 in i-butyl methyl ether. The ethereal solution was evaporated to give 20 as a red-brown oil, in approximately quantitative yield. ¾-NMR (300 MHz, CDCb) ppm δ 7.05-7.03 (m, 2H), 6.70 (t, / = 7.5 Hz, 1H), 6.60 (d, / = 7.8Hz, 1H), 6.74 (s, 1H), 2.77 (bs, 2H), 2.64 (t, / = 7.2 Hz, 2H), 2.56 (t, / = 7.2 Hz, 2H), 1.69 (quint, / = 7.2 Hz, 2H); 13 C-NMR (75 MHz, CDCb) ppm δ 136.27, 127.11, 121.43, 121.16, 118.40, 118.38, 114.86, 111.08, 61.45, 41.40, 33.36, 29.61, 22.14, 13.86.

Measurement of protective activity against oxidative stress in a macrophage cell culture

Compounds of the invention were tested for their potential to protect against cell death induced by oxidative stress in a mouse macrophage (RAW 264.7) cell line. Cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM), 4500 mg/L D-glucose with 10% foetal calf serum (FCS), 10000 U/mL penicillin, 100 mg/mL streptomycin and 25 μg/mL Amphotericin B at 37°C with 95% air and 5% CO2. Cells were seeded in 96 well- plates at a density of lxlO 4 cells/well and incubated with different compounds of the invention in several concentrations ranging from 10 "14 -10 "9 M, for 2 h prior to addition of H2O2 (150 μΜ). Cell viability was measured 24 h later by means of the MTT (3-(4,5- dimethyl thiazol-2-yl)-2,5,diphenyl tetrazolium bromide) assay. MTT measures the activity of mitochondrial dehydrogenase in viable cells and is based on the reduction of yellow MTT to dark blue formazan crystals by mitochondrial dehydrogenases (succinate dehydrogenase). MTT solution was added to each well in a final concentration of 0.5 mg/ml. After 2 h, the MTT containing medium was aspired. Cells were lysed with 3% SDS and formazan crystals were dissolved in isopropanol/HCl. Optical density was determined by means of a plate-reader at a wavelength of 570 nm. Cell viability was reduced by H2O2 by 25-35% relative to that in control cells.

Measurement of caspase 3 activity in RAW macrophage 264.7 cells

Oxidative stress can cause apoptosis, a form of cell suicide mediated by a cascade of proteolytic enzymes called caspases (cysteinyl aspartate- specific proteases). Caspase 3 is one of a number of effector caspases. Its activity was measured by means of a luminescent assay (Caspase-Glo 3/7 Assay Promega Ltd). Macrophages were pre-treated as described above with concentrations of 10 "12 and 10 "9 M of several of the compounds 4 h before the addition of H2O2 (100 μΜ). The medium was aspirated 90 min later, 100 μΐ, of DMEM was added to each well and the plates kept in an incubator overnight. Caspase-GloR Reagent (100 ί) was added to each well and the contents gently mixed using a plate shaker at 300-500 rpm for 30 sec. The plates were left at room temperature for 30 min and then luminescence of each sample was measured in a plate -reading luminometer (Cytation 3).

Measurement of anti-inflammatory activity in a mouse macrophage cell line culture

Anti-inflammatory activity was measured in RAW 264.7 macrophage cells. The cells were seeded at density of 5 x 10 4 per well in 48-well culture plates and grown overnight in DMEM as described above. The compounds were added to give final concentrations ranging from 1 x 10 "12 -1 x 10 "6 M of the respective salts per well. The cells were incubated for 2 h at 37 °C prior to stimulation with lipopolysaccharide (LPS, 2.5 μg/mL) from Escherichia coli (Sigma- Aldrich). Supernatants were harvested after 8 h for detection of TNF-a and after 24 h for nitric oxide (NO). NO production was detected by a colourimetric method using Griess reagent (0.2% naphthylenediamine dihydrochloride, and 2% sulfanilamide in 5% phosphoric acid), which measures the concentration of nitrite, a stable metabolite produced from NO. TNF-a protein was detected by means of the sandwich ELISA method using an ELISA kit (Biolegend, San Diego, CA, USA) according to the manufacturer's instructions.

Measurement of anti-inflammatory activity in human monocytes and macrophages

Blood (20 ml was collected from healthy human subjects on the day of the experiment in anti-coagulant tubes and diluted in PBS 1 :1. Ficol (12 mL) was added into the bottom of the tube, centrifuged slowly for 30 min at 1400 g and peripheral blood mononuclear cells (PBMCs) were separated. Monocytes were isolated by addition of 15 ml of MACS buffer. After centrifugation at 1400 g for 10 min at 4 °C, the supernatant fluid was removed, monocytes were isolated by means of a MACS-Monocyte isolation kit (cat: 130-091-153) and incubated at 4-8 °C for 10 min. Monocytes (500,000/well) were seeded in 12 multi well plate and cultured for 7 days in RPMI + L-Glutamin + FBS (10%) and PenStrep and h-MCSF (20 ng/mL) to convert them into macrophages. Compounds (AN1284, AN1297, AN1298, lxlO 10 or lxlO "9 M) were added 2 h before LPS (100 μg/mL Sigma Ltd. phenol extracted from Escherichia coli 055:B5) and incubated at 37 °C, 5% C0 2 . TNF-a, IL-Ιβ and IL-6 protein were detected by means of the sandwich ELISA method using an ELISA kit for human cytokines (Biolegend, San Diego, CA, USA) according to the manufacturer's instructions.

Evaluation of anti-inflammatory activity in mice

Male Balb/c OlaHsd mice (aged 7-8 weeks) were injected subcutaneously (sc) with saline, or with AN1279, AN1283, AN1284, AN1287 and AN1298 at doses that contain approximately the equivalent amount of the base. LPS (10 mg/kg) was given by intraperitoneal injection 15 min later. The mice were sacrificed 4 h later, which was previously shown to be the optimal time for elevation of cytokines in the spleen and brain. The spleen, brain and liver were rapidly removed and frozen in liquid nitrogen and stored at -80 °C until use. A piece of the cortex was taken for gene analysis of cytokines. Blood was collected into heparinized tubes and plasma separated by centrifugation. For measurement of cytokine protein, tissues were weighed and diluted in PBS containing 0.8% NaCl, 0.144% NaHP0 4 , 0.024% KH 2 P0 4 and protease inhibitor cocktail (Sigma) and homogenised in an Ultra-TURRAX® homogeniser at a speed of 24,000 rpm and centrifuged at 14000g for 15 min at 4 °C. Cytokines were detected by means of Elisa kits (Biolegend) and protein concentration was determined by a bicinchoninic acid protein assay kit (Thermo Scientific, IL, USA).

For evaluation of cytokine mRNA, RNA was extracted from the tissue with Tri Reagent® (Sigma) and was reverse transcribed into cDNA by means of a high capacity cDNA reverse transcription kit (Applied Biosy stems). For qRT-PCR, TaqMan Fast Universal PCR Master Mix (Applied Biosystems) and TaqMan Gene Expression Assays reagents (Applied Biosystems) were used. Hypoxanthine phosphoribosyl transferase (HPRT) was used as house-keeping gene and all results are normalised thereto.

Pharmacodynamic assessment of indoline derivatives for tuberous sclerosis-

RERT/TSCl f/f mice develop kidney pathology that resembles human tuberous sclerosis (TSC). Mice bearing a floxed allele of TSC1 were purchased from Jackson laboratories and crossed to homozygosity to the RERT strain, provided by Dr. Mariano Barbacid, Spanish National Cancer Research Centre. The RERT strain contains an IRES- Cre-ERT2 knock in into the large subunit of RNA polymerase II, allowing an inducible deletion in all cell types by administration of tamoxifen. Mice were kept on the mixed background originally provided by Jackson and bred for over 20 generations to form a congenic colony of RERT/TSC 1 f/f mice with an undefined genetic background that shows an accelerated development of disease. The mice were injected sc with tamoxifen to induce TSC1 deletion and its effect assessed on body weight and immunological parameters associated with inflammation. AN1284 (1 μπιοΐε^ of the 2HC1 salt) was injected sc, twice daily in a group of 5 female mice 5 weeks after tamoxifen challenge. An untreated group of 5 tamoxifen challenged mice served as controls. The mice were weighed once weekly and observed for development of lethargy and eye closure. Pharmacodynamic assessment of indoline derivatives for diabetic nephropathy

The renal proximal tubule is uniquely susceptible to a variety of metabolic and hemodynamic factors associated with diabetes, predominantly to hyperglycaemia. Glucose entry into renal proximal tubular cells is insulin-independent, making these cells particularly sensitive to the deleterious effects of chronic hyperglycaemia in subjects with diabetes. To induce diabetic nephropathy, male 20, 8-week-old C57B1/6 mice were given five consecutive intraperitoneal injections of streptozotocin (STZ) (50 mg/kg per day). A control group of 10 mice was given 0.1 mol/L citrate buffer (pH 4.5). Ten days following the last injection of STZ, half the mice were treated twice daily with sc injections of AN1284 2HC1 (2 umoles/kg), or vehicle (saline) for fifteen weeks.

Pharmacodynamic assessment of indoline derivatives for acute pancreatitis

The most commonly used animal model for acute pancreatitis is the cerulein- induced mouse model (Su et al., 2006). Cerulein is a cholecystokinin analogue that produces hyperstimulation of the pancreatic acinar cells at supramaximal dosage. The acute necrotizing pancreatitis produced is histopathologically similar to acute human pancreatitis (Van Acker et al., 2007).

Groups of 10 Balb/C male mice weighing 24 gm were injected intraperitoneally once every hour for 4 hours with cerulean (50 μg/kg) suspended in saline. AN 1297 (1 or 2 mg/kg) or resolvin D (1 mg/kg) or saline that were injected sc. 30 min after the first cerulean injection. Mice were killed by exsanguination at 4 hours after the first cerulein injection. Blood samples or measurement of amylase were obtained by direct intracardiac puncture. Pancreases were removed immediately, flash frozen in liquid nitrogen and stored -80°C until assayed, for TNF-a protein by means of the sandwich ELISA method using an ELISA kit (Biolegend, San Diego, CA, USA) according to the manufacturer's instructions. Serum amylase activity was determined using a colorimetric assay kit (abl02523, Abeam). The values of serum amylase activity are expressed as units per litre (U/l).

RESULTS

Compounds AN1283, AN1284, AN1287, AN1292, AN1294, AN1295, AN1296, AN1297, AN1298, AN1299 and AN1400, were tested as HC1 or 2HC1 (AN1298) salts and the proportion of base in the total salt ranged from 0.78-0.88. Compounds AN1285, AN 1279 and AN 1293 were tosylate salts in which the proportion of base in the total salt ranged from 0.57-0.63. Compounds AN1264, AN1276, AN1282 were di-tosylate salts in which the proportion of base in the total salt ranged from 0.22-0.37.

Protection activity of compounds of the invention against oxidative stress in cell cultures

Significant protection (increase in viability to level of controls without H 2 O 2 ) was obtained over the concentration range of lxlO "13 - lxlO "9 M by the following compounds; AN1264, AN1282, AN1284, AN1285, AN1287, AN1292, AN1297 and AN1299. AN1283, AN1276 and AN1279 caused this level of protection at concentrations of 1x10 " 13 -lxl0 "n M and compounds AN647, AN1287 and AN1400, at concentrations of 1x10 " 10 - lxlO "9 M.

Reduction by compounds in elevation of caspase 3 induced in RAW macrophages

All the compounds tested at concentrations of 10 "13 and 10 "9 M significantly reduced caspase 3 activity in macrophages subjected to oxidative stress with H2O2 (Fig. 1). AN1264 and AN1279 (lxlO 13 M) caused less reduction in caspase 3 activity than the other compounds in keeping with their smaller degree of protection in the MTT assay.

Reduction by the compounds of NO and cytokines, TNF-a and IL-6 in LPS- stimulated macrophages

The steroid budesonide was used as a positive control for these experiments. The range of concentrations with which a reduction of at least 25% in NO release from LPS- stimulated macrophages was obtained (ranging up to 50% at concentrations of 1 nM) is shown in Table 1. Compounds that caused a significant decrease at the lowest concentrations of 1 x lO 12 or 1 x 10 "11 M were AN1283, AN1284 and AN1298. These compounds all have their side chains in position 1. Table 1. Reduction by compounds of release of NO from LPS-activated RAW cells

Data represent range of concentrations over which budesonide and the indoline derivatives significantly reduced the release of NO from LPS activated RAW macrophages.

Table 2. Reduction by compounds of release of TNF-a from LPS-activated RAW cells

Data represent range of concentrations over which budesonide and the indoline derivatives significantly reduced the release of TNF-a from LPS activated RAW macrophages.

They were also the most potent as inhibitors of the release of TNF-a (Table 2). When the propionic ester or propylamine side chain was in position 3 (AN647 and AN1282, respectively) the lowest concentration that inhibited NO and TNF-a release by 25% was 1 x 10 "10 M. Substitution of propylamine by N-isopropylpropylamine (AN 1284) decreased by 10 fold the concentration that inhibited significantly the release of NO and TNF-a. While introduction of OCH 3 in position 5 of AN1284 (AN1298) does not affect the activity of the compound, a 100-fold greater concentration of the amine with 5 -CI substituent (AN1285) was need to cause the same effect. Anti-inflammatory activity of compounds in human monocytes

The effect of AN1284, AN1297 and AN1298 on the release of TNF-a and IL-6 from human monocytes activated by LPS is shown in Table 1. TNF-a was measured 6, 12 and 24 h after addition of LPS in different subjects and that of IL-6 only after 24 h. All three compounds cause a similar, concentration-dependent statistically significant reduction in TNF-a, which was greatest 24 h after addition of LPS. They also cause a similar reduction of IL-6 as shown in Table 3.

Table 3. Reduction by three indoline derivatives of TNF-a and IL-6 in human macrophages after their elevation by LPS.

Time after Cytokine Concentration Reduction (%)± STD compared to LPS

LPS (h) M alone

Compounds

AN1284 AN1297 AN1298

TNF-a

6 1 x lO -9 55.3 + 9.8 55.7 + 11.5 50.4 + 13.5

12 1 x 10- 11 7.1 + 5.8* 22.0 + 7.2 14.6 + 4.2

12 1 x It) "10 41.1 + 5.4 25.1 + 10.9 28.0 + 7.9

24 1 x lO -9 71.4 + 9.7 71.1 + 8.9 72.2 + 9.7

IL-6

24 1 x 10 -9 64.7 + 14.8 62.4 + 16.5 54.6 + 18.8

All reductions except that marked by # are significant /?<0.05.

Reduction of pro-inflammatory cytokine proteins in tissues of mice injected with LPS

The only cytokine protein that increases consistently in the brain and liver to measurable amounts, 4 h after LPS injection was IL-6. Figure 2 shows the reduction by 2-3 doses (expressed as the μπιο^^ of the base) of AN1283, AN1284, AN1297 and AN1298 of IL-6 in the brain and liver. Both IL-6 and TNF-a increased in plasma and spleen after LPS. The optimal time for elevation of these cytokines in plasma is 1.5-2 h. The reduction of these cytokines by different doses of four AN compounds is shown in Figures 3 and 4 respectively. Reduction of pro-inflammatory cytokine mRNAs in brain of mice injected with LPS

Four compounds also reduce the expression of pro-inflammatory genes, TNF-a, IL-Ιβ and IL-12b in the brain that are increased by injection of LPS (Figure 5). This shows that the compounds are able to reach the CNS.

Reduction of morbidity and immune parameters in TSCl-deleted mice

Signs of deteriorating health were observed within 8-10 weeks following tamoxifen challenge in the mice. This was manifested by reduction in body weight, lethargic behaviour and the appearance of eye exudates and seizures, a pathology associated with the human disease. Ten weeks from the time of tamoxifen injection, mice were sacrificed and analysed for gross pathology and markers of inflammation. The kidneys were enlarged with the development of large cysts (Fig. 6A). Staining with Ki67, a marker for proliferating cells revealed massive proliferation in the TSCl KO mice indicative of development of a benign tumour or hyper-proliferation (hamartomas) that are typical for TSC (Fig. 6B).

Five weeks after the initiation of AN 1284 treatment the untreated group lost much weight and were so lethargic that they had to be terminated. Remarkably, none of the AN1284-treated mice developed severe lethargy and their body weight showed a much smaller decrease (Fig. 7). Flow cytometry analyses revealed a significant reduction in MDSCs in the bone marrow and spleen and in T cells in the bone marrow and spleen in the AN1284-treated mice (Fig. 8). A significant reduction was also shown for cytokine levels of TNF- a and IL-6 measurements in spleen and liver in the AN- 1284 treated mice as compared with the untreated group (Fig. 9).

The kidneys of AN1284-treated mice showed a clear reduction in renal cysts and retention of the aligning columnar epithelial cells in the cyst, while these were missing in those of the mice treated only with tamoxifen. There was also a 50-60% reduction in the number of Ki67-positive cells proliferating cells following drug treatment. The data indicate that the treatment with AN 1284 is effective in reducing the kidney pathology associated with TSCl deletion in the RERT/TSCl f/f model even when initiated at a late stage (Fig. 10). Reduction of renal damage in type 1 diabetic mice with renal nephropathy-deleted mice

Compared with the controls, the diabetic mice did not gain appreciable body weight during the study (Fig. 11 A). The reduction in body weight was due to a decrease in total fat mass (Fig. 11B) with no change in lean body mass (Fig. 11C). Interestingly, AN1284 induced a further reduction in total fat mass. AN1284 did not affect the degree of hyperglycaemia (Fig. 1 ID). The kidney-to-body weight ratio which increases in the diabetic-vehicle (Veh)-treated group was significantly reduced by AN1284. Moreover, chronic treatment with AN1284 was effective in normalizing urine albumin-to-creatinine ratio (Fig. 12A) and blood urea nitrogen levels (Fig. 12B), glomerular cross-sectional area (Fig. 13). Compared with nondiabetic control mice, increased glomerular space area and mesangial expansion were noted in the diabetic-vehicle-treated animals, effects that were completely normalised by AN 1284 (Fig. 14A-14B). AN1284 also significantly reduced the elevated mRNA expression levels of the kidney injury marker, lipocalin 2, in whole kidney homogenates of diabetic mice (Fig. 15 A) and those of collagen- 1 (Fig 15B), collagen-3 (Fig. l5C), TIMP1 (Fig. l5D) and IP-10 (Fig. 15E). These data indicate a robust efficacy of AN1284 for DN in a manner independent of the diabetes itself.

Reduction of acute pancreatitis induced by cerulein in mice.

Serum amylase increased 7-fold, following cerulein injections. These were decreased significantly by AN1297 (1 and 2 mg/kg) in a dose dependent manner (Fig. 16A). AN1297 also dose-dependently reduced TNF-a protein in the pancreas (Fig. 16B).