Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENT AND PREVENTION OF ALCOHOLIC LIVER DISEASE
Document Type and Number:
WIPO Patent Application WO/2023/006765
Kind Code:
A1
Abstract:
Methods of treating or preventing alcoholic liver disease are disclosed, the methods comprising administering to a subject a therapeutically or prophylactically effective amount of an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling.

Inventors:
EFFENBERGER MARIA (AT)
TILG HERBERT (AT)
Application Number:
PCT/EP2022/070974
Publication Date:
February 02, 2023
Filing Date:
July 26, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOEHRINGER INGELHEIM INT (DE)
International Classes:
A61P1/16; C07K14/54; C07K14/715; C07K16/24
Domestic Patent References:
WO2020225147A12020-11-12
WO1999059608A21999-11-25
WO2018109174A22018-06-21
WO2019238882A12019-12-19
WO2014121325A12014-08-14
WO2018109170A22018-06-21
WO2019238884A12019-12-19
WO2017103108A12017-06-22
WO2018109168A12018-06-21
WO2009052588A12009-04-30
WO1991019813A11991-12-26
WO2001029058A12001-04-26
WO1999032619A11999-07-01
Foreign References:
US20200262910A12020-08-20
GB202110721A2021-07-26
GB202110862A2021-07-28
US20090202533A12009-08-13
US20140219919A12014-08-07
US20130302277A12013-11-14
US6649192B22003-11-18
US5843509A1998-12-01
Other References:
COOK STUART A ET AL: "Hiding in Plain Sight: Interleukin-11 Emerges as a Master Regulator of Fibrosis, Tissue Integrity, and Stromal Inflammation", ANNU. REV. MED, 1 January 2020 (2020-01-01), pages 263 - 76, XP055975091, Retrieved from the Internet [retrieved on 20221026], DOI: 10.1146/annurev-med-041818-
MIYAWAKI AKIMITSU ET AL: "IL-11 prevents IFN-[gamma]-induced hepatocyte death through selective downregulation of IFN-[gamma]/STAT1 signaling and ROS scavenging", PLOS ONE, vol. 14, no. 2, 19 February 2019 (2019-02-19), pages e0211123, XP055975065, Retrieved from the Internet DOI: 10.1371/journal.pone.0211123
UNDERHILL-DAY N ET AL: "FUNCTIONAL CHARACTERIZATION OF W147A: A HIGH-AFFINITY INTERLEUKIN-11 ANTAGONIST", ENDOCRINOLOGY, THE ENDOCRINE SOCIETY, US, vol. 144, no. 8, 1 August 2003 (2003-08-01), pages 3406 - 3414, XP009016241, ISSN: 0013-7227, DOI: 10.1210/EN.2002-0144
LANCET, vol. 392, 2018, pages 1015 - 1035
ASRANI ET AL., J HEPATOL, vol. 70, 2019, pages 151 - 171
AVILA ET AL., GUT, vol. 69, 2020, pages 764 - 780
LANG ET AL., GUT MICROBES, vol. 12, 2020, pages 1785251
PARLESAK ET AL., J HEPATOL, vol. 32, 2000, pages 742 - 747
TILGDIEHL, N ENGL J MED, vol. 343, 2000, pages 1467 - 1476
LOPETUSO ET AL., LIVER. INT J MOL SCI, 2018, pages 19
SCHMIDT-ARRASROSE-JOHN, J HEPATOL, vol. 64, 2016, pages 1403 - 1415
BARBIER ET AL., FRONT IMMUNOL, vol. 10, 2019, pages 2014
WIDJAJA ET AL., GASTROENTEROLOGY, vol. 157, no. 3, 2019, pages 777 - 792
CURTIS ET AL., BLOOD, vol. 90, no. 11, 1997, pages 4403 - 3908
GARBERSSCHELLER, BIOL. CHEM., vol. 394, no. 9, 2013, pages 1145 - 1161
"UniProt", Database accession no. POCG04-1
KARPOVICH ET AL., MOL. HUM. REPROD., vol. 9, no. 2, 2003, pages 75 - 80
METCALFE ET AL.: "Manuscript RA119.012351", JBC, 2020
HILTON ET AL., EMBO J, vol. 13, 1994, pages 4765
NANDURKAR ET AL., ONCOGENE, vol. 12, 1996, pages 585
PFLANZ, FEBS LETT, vol. 450, 1999, pages 117 - 122
CURTIS ET AL., BLOOD, vol. 90, no. 11, 1 December 1997 (1997-12-01), pages 4403 - 12
LOKAU ET AL., CELL, vol. 14, 2016, pages 1761 - 1773
PUTOCZKIERNST, J LEUKOC BIOL, vol. 88, no. 6, 2010, pages 1109 - 1117
SEITZ, NATURE REVIEWS DISEASE PRIMERS, vol. 4, 2018, pages 16
SEITZ ET AL., J. CLIN. MED., vol. 10, 2021, pages 858
OSNA ET AL., ALCOHOL RES., vol. 38, no. 2, 2017, pages 147 - 161
TORRUELLAS ET AL., WORLD J GASTROENTEROL., vol. 20, no. 33, 2014, pages 11684 - 11699
ALTENHOFER, ANTIOXID REDOX SIGNAL., vol. 23, no. 5, 2015, pages 406 - 427
AUGSBURDER, REDOX BIOL., vol. 26, 2019, pages 101272
NEUBERGER ET AL.: "Monoclonal Antibodies: A manual of techniques", 1988, CRC PRESS, pages: 792 - 799
J G R HURRELL: "Monoclonal Hybridoma Antibodies: Techniques and Applications", 1982, CRC PRESS
MORRISON ET AL., PROC. NATL. ACAD. SD. USA, vol. 81, 1984, pages 6851 - 6855
BOCKHORN ET AL., NAT. COMMUN., vol. 4, no. 0, 2013, pages 1393
HERMANN ET AL., ARTHRITIS RHEUM., vol. 41, no. 8, 1998, pages 1388 - 97
NG ET AL., SCI TRANSL MED, vol. 11, no. 511, 2019, pages eaaw1237
SCHAEFER ET AL., NATURE, vol. 552, no. 7683, 2017, pages 110 - 115
NG ET AL.: "IL-11 is a therapeutic target in idiopathic pulmonary fibrosis", BIORXIV 336537; DOI: HTTPS://DOI.ORG/10.1101/336537
BLANC ET AL., J. IMMUNOL METHODS., vol. 241, no. 1-2, 31 July 2000 (2000-07-31), pages 43 - 59
WIDJAJA ET AL.: "IL-11 neutralising therapies target hepatic stellate cell-induced liver inflammation and fibrosis in NASH", BIORXIV 470062; DOI: HTTPS://DOI.ORG/10.1101/470062
PARKSMOLEN, ADVANCES IN PROTEIN CHEMISTRY, vol. 56, 2001, pages 369 - 421
ALMAGROFRANSSON, FRONTIERS IN BIOSCIENCE, vol. 13, 2008, pages 1619 - 1633
SAFDARI ET AL., BIOTECHNOLOGY AND GENETIC ENGINEERING REVIEWS, vol. 29, no. 2, 2013, pages 175 - 186
LO ET AL., MICROBIOLOGY SPECTRUM, vol. 2, no. 1, 2014
BRIIGGEMANN ET AL., ARCH IMMUNOL THER EXP (WARSZ, vol. 63, no. 2, 2015, pages 101 - 108
HOOGENBOOM, NAT. BIOTECHNOL., vol. 23, 2005, pages 1105 - 1116
CHAN ET AL., INTERNATIONAL IMMUNOLOGY, vol. 26, no. 12, 2014, pages 649 - 657
NORDAN, R. P., J. IMMUNOL., vol. 139, 1987, pages 813
NG ET AL., SCI TRANSL MED., vol. 11, no. 511, 2019, pages eaaw1237
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
CHOTHIA, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
RETTER ET AL., NUCL. ACIDS RES., vol. 33, 2005, pages D671 - D674
DAVIESSUTTON, IMMUNOL REV., vol. 268, no. 1, November 2015 (2015-11-01), pages 139 - 159
ANGAL ET AL., MOL IMMUNOL., vol. 30, no. 1, January 1993 (1993-01-01), pages 105 - 8
MARKS ET AL., RIOLTECHNOLOGY, vol. 10, 1992, pages 779 - 783
BARBAS ET AL., PROC NAT. ACAD. SCI. USA, vol. 91, 1994, pages 3809 - 3813
SCHIER, GENE, vol. 169, 1995, pages 147 - 155
JACKSON, J. IMMUNOL., vol. 154, no. 7, 1995, pages 331 0 - 15 9
HAWKINS ET AL., J. MOL. BIOL., vol. 226, 1992, pages 889 - 896
MUELLER, D ET AL., BIODRUGS, vol. 24, no. 2, 2010, pages 89 - 98
WOZNIAK-KNOPP G ET AL., PROTEIN ENG DES, vol. 23, no. 4, 2010, pages 289 - 297
BAEUERLE, PA ET AL., CANCER RES, vol. 69, no. 12, 2009, pages 4941 - 4944
KONTERMANN, MABS, vol. 4, no. 2, 2012, pages 182 - 197
D. M.BAST, B. J.: "Production of Bispecific Antibodies", CURRENT PROTOCOLS IN IMMUNOLOGY, vol. 14, no. IV, 2001, pages 1 - 16
DE-CHAO YU ET AL., MOLECULAR THERAPY, vol. 20, no. 5, 2012, pages 938 - 947
HORNIGFARBER-SCHWARZ: "French, How to make bispecific antibodies", METHODS MOL. MED., vol. 40, 2000, pages 333 - 339
KONNERDUPONT, CLIN COLORECTAL CANCER, vol. 4, October 2004 (2004-10-01), pages 81 - 5
UNDERHILL-DAY, ENDOCRINOLOGY, vol. 144, no. 8, August 2003 (2003-08-01), pages 3406 - 14
LEE ET AL., AM J RESPIRE CELL MOL BIOL., vol. 39, no. 6, December 2008 (2008-12-01), pages 739 - 746
MENKHORST ET AL., BIOLOGY OF REPRODUCTION, vol. 80, no. 5, 1 May 2009 (2009-05-01), pages 920 - 927
PASQUALINI, CANCER, vol. 121, no. 14, 2015, pages 2411 - 2421
LAY ET AL., INT. J. ONCOL., vol. 41, no. 2, 2012, pages 759 - 764
GOLD L ET AL., PLOS ONE, vol. 5, no. 12, 2010, pages e15004
TUERKGOLD, SCIENCE, vol. 249, no. 4968, 1990, pages 505 - 10
SINHA, N. D.BIERNAT, J.MCMANUS, J.KOSTER, H., NUCLEIC ACIDS RES., vol. 12, 1984, pages 4539
BEAUCAGE, S. L.LYER, R. P., TETRAHEDRON, vol. 48, no. 12, 1992, pages 2223
REVERDATTO ET AL., CURR TOP MED CHEM., vol. 15, no. 12, 2015, pages 1082 - 101
HEARTY ET AL., METHODS MOL BIOL, vol. 907, 2012, pages 411 - 442
RICH ET AL., ANAL BIOCHEM., vol. 373, no. 1, 1 February 2008 (2008-02-01), pages 112 - 20
LAD ET AL., J BIOMOL SCREEN, vol. 20, no. 4, 2015, pages 498 - 507
CONCEPCION ET AL., COMB CHEM HIGH THROUGHPUT SCREEN., vol. 12, no. 8, September 2009 (2009-09-01), pages 791 - 800
JERABEK-WILLEMSEN ET AL., ASSAY DRUG DEV TECHNOL., vol. 9, no. 4, August 2011 (2011-08-01), pages 342 - 353
"Springer Protocols", vol. 1, 2010, SPRINGER, article "Antibody Engineering", pages: 657 - 665
TAKI, CLIN EXP IMMUNOL, vol. 112, no. 1, 1998, pages 133 - 138
"GenBank", Database accession no. NM_001008725.3
NATURE, vol. 457, no. 7228, 22 January 2009 (2009-01-22), pages 426 - 433
JOHN ET AL., PLOS BIOLOGY, vol. 11, no. 2, 2004, pages 1862 - 1879
MYERS, NATURE BIOTECHNOLOGY, vol. 21, 2003, pages 324 - 328
SHINAGAWA ET AL., GENES AND DEV., vol. 17, 2003, pages 1340 - 5
SHEN ET AL., FEBS LETT, vol. 539, no. 1-3, 27 March 2003 (2003-03-27), pages 111 - 4
BARTONMEDZHITOV, PNAS, vol. 99, no. 23, 12 November 2002 (2002-11-12), pages 14943 - 14945
WANG ET AL., AAPS J., vol. 12, no. 4, December 2010 (2010-12-01), pages 492 - 503
FIRE A ET AL., NATURE, vol. 391, 1998, pages 806 - 811
FIRE, A., TRENDS GENET., vol. 15, 1999, pages 358 - 363
SHARP, P. A.: "RNA interference", GENES DEV., vol. 15, 2001, pages 485 - 490, XP002235698, DOI: 10.1101/gad.880001
HAMMOND, S. M. ET AL., NATURE REV. GENET., vol. 2, 2001, pages 110 - 1119
TUSCHL, T., CHEM. BIOCHEM., vol. 2, 2001, pages 239 - 245
HAMILTON, A ET AL., SCIENCE, vol. 286, 1999, pages 950 - 952
HAMMOND, S. M. ET AL., NATURE, vol. 404, 2000, pages 293 - 296
ZAMORE, P. D. ET AL., CELL, vol. 101, 2000, pages 25 - 33
BERNSTEIN, E. ET AL., NATURE, vol. 411, 2001, pages 494 - 498
ELBASHIR, S. M. ET AL., GENES DEV., vol. 15, 2001, pages 188 - 200
SHIN, RNA, vol. 15, no. 5, May 2009 (2009-05-01), pages 898 - 910
DZAU, TRENDS IN BIOTECHNOLOGY, vol. 11, 2003, pages 205 - 210
BORKHARDT, A.: "Blocking oncogenes in malignant cells by RNA interference--new hope for a highly specific cancer treatment?", CANCER CELL, vol. 2, 2002, pages 167 - 8, XP009006463, DOI: 10.1016/S1535-6108(02)00129-0
HANNON, G.J.: "RNA interference", NATURE, vol. 418, 2002, pages 244 - 51, XP002979088, DOI: 10.1038/418244a
MCMANUS, M.T.P.A. SHARP: "Gene silencing in mammals by small interfering RNAs", NAT REV GENET., vol. 3, 2002, pages 737 - 47, XP002352198, DOI: 10.1038/nrg908
SCHERR, M.M.A. MORGANM. EDER.: "Gene silencing mediated by small interfering RNAs in mammalian cells", CURR MED CHEM., vol. 10, 2003, pages 245 - 56, XP009019025
SHUEY, D.J.D.E. MCCALLUST. GIORDANO: "RNAi: gene-silencing in therapeutic intervention", DRUG DISCOV TODAY, vol. 7, 2002, pages 1040 - 6, XP002331644, DOI: 10.1016/S1359-6446(02)02474-1
LEWIS, D.L., J.E. HAGSTROM, A.G. LOOMIS, J.A. WOLFF, H. HERWEIJER: "Efficient delivery of siRNA for inhibition of gene expression in postnatal mice", NAT GENET, vol. 32, 2002, pages 107 - 8, XP002978928, DOI: 10.1038/ng944
PAUL, C.P.P.D. GOODI. WINERD.R. ENGELKE: "Effective expression of small interfering RNA in human cells", NAT BIOTECHNOL., vol. 20, 2002, pages 505 - 8, XP001121066, DOI: 10.1038/nbt0502-505
SONG, E.S.K. LEEJ. WANGN. INCEN. OUYANGJ. MINJ. CHENP. SHANKARJ. LIEBERMAN: "RNA interference targeting Fas protects mice from fulminant hepatitis", NAT MED, vol. 9, 2003, pages 347 - 51, XP002351920, DOI: 10.1038/nm828
SORENSEN, D.R.M. LEIRDALM. SIOUD: "Gene silencing by systemic delivery of synthetic siRNAs in adult mice", J MOL BIOL., vol. 327, 2003, pages 761 - 6, XP027101312, DOI: 10.1016/S0022-2836(03)00181-5
ABBAS-TERKI, T.W. BLANCO-BOSEN. DEGLONW. PRALONGP. AEBISCHER: "Lentiviral-mediated RNA interference", HUM GENE THER, vol. 13, 2002, pages 2197 - 201, XP002978528, DOI: 10.1089/104303402320987888
BARTON, G.M.R. MEDZHITOV.: "Retroviral delivery of small interfering RNA into primary cells", PROC NATL ACAD SCI USA., vol. 99, 2002, pages 14943 - 5, XP002328325, DOI: 10.1073/pnas.242594499
DEVROE, E.P.A. SILVER.: "Retrovirus-delivered siRNA", BMC BIOTECHNOL, vol. 2, 2002, pages 15, XP021017015, DOI: 10.1186/1472-6750-2-15
LORI, F.P. GUALLINIL. GALLUZZIJ. LISZIEWICZ: "Gene therapy approaches to HIV infection", AM J PHARMACOGENOMICS, vol. 2, 2002, pages 245 - 52
MATTA, H.B. HOZAYEVR. TOMARP. CHUGHP.M. CHAUDHARY: "Use of lentiviral vectors for delivery of small interfering RNA", CANCER BIOL THER, vol. 2, 2003, pages 206 - 10, XP001180107
QIN, X.F.D.S. ANI.S. CHEND. BALTIMORE: "Inhibiting HIV-1 infection in human T cells by lentiviral-mediated delivery of small interfering RNA against CCR5", PROC NATL ACAD SCI USA., vol. 100, 2003, pages 183 - 8, XP002975768, DOI: 10.1073/pnas.232688199
SCHERR, M.K. BATTMERA. GANSERM. EDER: "Modulation of gene expression by lentiviral-mediated delivery of small interfering RNA", CELL CYCLE, vol. 2, 2003, pages 251 - 7
SHEN, C.A.K. BUCKX. LIUM. WINKLERS.N. RESKE: "Gene silencing by adenovirus-delivered siRNA", FEBS LETT, vol. 539, 2003, pages 111 - 4, XP004415445, DOI: 10.1016/S0014-5793(03)00209-6
MORRIS, M.C.L. CHALOINF. HEITZG. DIVITA: "Translocating peptides and proteins and their use for gene delivery", CURR OPIN BIOTECHNOL, vol. 11, 2000, pages 461 - 6, XP001022944, DOI: 10.1016/S0958-1669(00)00128-2
SIMEONI, F.M.C. MORRISF. HEITZG. DIVITA: "Insight into the mechanism of the peptide-based gene delivery system MPG: implications for delivery of siRNA into mammalian cells", NUCLEIC ACIDS RES., vol. 31, 2003, pages 2717 - 24, XP002984580, DOI: 10.1093/nar/gkg385
"UniProtKB", Database accession no. P20809
"Remington's Pharmaceutical Sciences", 1990, LIPPINCOTT, WILLIAMS & WILKINS
"Handbook of Pharmaceutical Excipients", 1994
PETER STARKEL: "Gut", vol. 57, 2008, pages: 440 - 442
WRIGHT ET AL., MOL. CELL. BIOL., vol. 30, no. 6, March 2010 (2010-03-01), pages 1411 - 1420
ANTHONY J. BROOKES: "The essence of SNPs", GENE, vol. 234, 8 July 1999 (1999-07-08), pages 177 - 186
FAN ET AL.: "Highly Parallel SNP Genotyping", COLD SPRING HARB SYMP QUANT BIOL, vol. 68, 2003, pages 69 - 78, XP001538040, DOI: 10.1101/sqb.2003.68.69
MATSUZAKI ET AL.: "Parallel Genotyping of Over 10,000 SNPs using a one-primer assay on a high-density oligonucleotide array", GENOME RES, vol. 14, 2004, pages 414 - 425, XP002430641, DOI: 10.1101/gr.2014904
SODING, J, BIOINFORMATICS, vol. 21, 2005, pages 951 - 960
NOTREDAME, J. MOL. BIOL., vol. 302, 2000, pages 205 - 217
LASSMANNSONNHAMMER, BMC BIOINFORMATICS, vol. 6, no. 298, 2005
KATOHSTANDLEY, MOLECULAR BIOLOGY AND EVOLUTION, vol. 30, no. 4, 2013, pages 772 - 780
SAMBROOK, J.RUSSEL, D.W.: "Molecular Cloning, A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
BERTOLA ET AL., NAT PROTOC, vol. 8, 2013, pages 627 - 637
GAO ET AL., GASTROENTEROLOGY, vol. 141, 2011, pages 1572 - 1585
MANDREKAR, HEPATOLOGY, vol. 64, 2016, pages 1343 - 1355
SEITZ ET AL., NAT REV DIS PRIMERS, vol. 4, 2018, pages 16
LOUVET ET AL., NAT REV GASTROENTEROL HEPATOL, vol. 12, 2015, pages 231 - 242
Attorney, Agent or Firm:
MEWBURN ELLIS LLP (GB)
Download PDF:
Claims:
Claims:

1. An agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling for use in a method of treating or preventing alcoholic liver disease.

2. Use of an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling in the manufacture of a medicament for use in a method of treating or preventing alcoholic liver disease.

3. A method of treating or preventing alcoholic liver disease, comprising administering a therapeutically or prophylactically effective amount of an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling to a subject.

4. The agent for use according to claim 1 , the use according to claim 2, or the method according to claim 3, wherein the agent is an agent capable of preventing or reducing the binding of interleukin 11 (IL-11) to a receptor for interleukin 11 (IL-11 R).

5. The agent for use according to claim 1 or claim 4, the use according to claim 2 or claim 4, or the method according to claim 3 or claim 4, wherein the agent is capable of binding to interleukin 11 (IL- 11) or a receptor for interleukin 11 (IL-11 R).

6. The agent for use according to any one of claims 1 , 4 or 5, the use according to any one of claims 2, 4 or 5, or the method according to any one of claims 3 to 5, wherein the agent is selected from the group consisting of: an antibody or an antigen-binding fragment thereof, a polypeptide, a peptide, a nucleic acid, an oligonucleotide, an aptamer or a small molecule.

7. The agent for use, the use or the method according to claim 5 or claim 6, wherein the agent is an antibody or an antigen-binding fragment thereof.

8. The agent for use, the use or the method according to claim 7, wherein the agent is an anti-IL-11 antibody antagonist of IL-11 -mediated signalling, or an antigen-binding fragment thereof.

9. The agent for use, the use or the method according to claim 7 or claim 8, wherein the antibody or antigen-binding fragment comprises:

(i) a heavy chain variable (VH) region incorporating the following CDRs:

HC-CDR1 having the amino acid sequence of SEQ ID NO:34 HC-CDR2 having the amino acid sequence of SEQ ID NO:35 HC-CDR3 having the amino acid sequence of SEQ ID NO:36; and

(ii) a light chain variable (VL) region incorporating the following CDRs:

LC-CDR1 having the amino acid sequence of SEQ ID NO:37 LC-CDR2 having the amino acid sequence of SEQ ID NO:38 LC-CDR3 having the amino acid sequence of SEQ ID NO:39.

10. The agent for use, the use or the method according to claim 7 or claim 8, wherein the antibody or antigen-binding fragment comprises:

(i) a heavy chain variable (VH) region incorporating the following CDRs:

HC-CDR1 having the amino acid sequence of SEQ ID NO:40 HC-CDR2 having the amino acid sequence of SEQ ID NO:41 HC-CDR3 having the amino acid sequence of SEQ ID NO:42; and

(ii) a light chain variable (VL) region incorporating the following CDRs:

LC-CDR1 having the amino acid sequence of SEQ ID NO:43 LC-CDR2 having the amino acid sequence of SEQ ID NO:44 LC-CDR3 having the amino acid sequence of SEQ ID NO:45.

11. The agent for use, the use or the method according to claim 7, wherein the agent is an anti-IL-11 Ra antibody antagonist of IL-11 -mediated signalling, or an antigen-binding fragment thereof.

12. The agent for use, the use or the method according to claim 7 or claim 11 , wherein the antibody or antigen-binding fragment comprises:

(i) a heavy chain variable (VH) region incorporating the following CDRs:

HC-CDR1 having the amino acid sequence of SEQ ID NO:46 HC-CDR2 having the amino acid sequence of SEQ ID NO:47 HC-CDR3 having the amino acid sequence of SEQ ID NO:48; and

(ii) a light chain variable (VL) region incorporating the following CDRs:

LC-CDR1 having the amino acid sequence of SEQ ID NO:49 LC-CDR2 having the amino acid sequence of SEQ ID NO:50 LC-CDR3 having the amino acid sequence of SEQ ID NO:51.

13. The agent for use, the use or the method according to claim 5 or claim 6, wherein the agent is a decoy receptor.

14. The agent for use, the use or the method according to claim 13, wherein the agent is a decoy receptor for IL-11.

15. The agent for use, the use or the method according to claim 14, wherein the decoy receptor for IL-11 comprises: (i) an amino acid sequence corresponding to the cytokine binding module of gp130 and (ii) an amino acid sequence corresponding to the cytokine binding module of IL-11 Ra.

16. The agent for use, the use or the method according to claim 5 or claim 6, wherein the agent is an IL- 11 mutein.

17. The agent for use, the use or the method according to claim 16, wherein the IL-11 mutein is W147A.

18. The agent for use according to claim 1, the use according to claim 2, or the method according to claim 3, wherein the agent is capable of preventing or reducing the expression of interleukin 11 (IL- 11) or a receptor for interleukin 11 (IL-11R).

19. The agent for use, the use, or the method according to claim 18, wherein the agent is an oligonucleotide or a small molecule.

20. The agent for use, the use or the method according to claim 19, wherein the agent is an antisense oligonucleotide capable of preventing or reducing the expression of IL-11.

21. The agent for use, the use or the method according to claim 20, wherein the antisense oligonucleotide capable of preventing or reducing the expression of IL-11 is siRNA targeted to IL11 comprising the sequence of SEQ ID NO:12, 13, 14 or 15.

22. The agent for use, the use or the method according to claim 19, wherein the agent is an antisense oligonucleotide capable of preventing or reducing the expression of IL-11Ra.

23. The agent for use, the use or the method according to claim 22, wherein the antisense oligonucleotide capable of preventing or reducing the expression of IL-11 Ra is siRNA targeted to IL11RA comprising the sequence of SEQ ID NO:16, 17, 18 or 19.

24. The agent for use, the use or the method according to any one of claims 4 to 23, wherein the interleukin 11 receptor is or comprises IL-11 Ra.

25. The agent for use according to any one of claims 1 , or 4 to 24, the use according to any one of claims 2, or 4 to 24, or the method according to any one of claims 3 to 24, wherein the method comprises administering the agent to a subject in which expression of interleukin 11 (IL-11) or a receptor for IL- 11 (IL-11 R) is upregulated.

26. The agent for use according to any one of claims 1 , or 4 to 25, the use according to any one of claims 2, or 4 to 25, or the method according to any one of claims 3 to 25, wherein the method comprises administering the agent to a subject in expression of interleukin 11 (IL-11) or a receptor for interleukin 11 (IL-11 R) has been determined to be upregulated.

27. The agent for use according to any one of claims 1 , or 4 to 26, the use according to any one of claims 2, or 4 to 26, or the method according to any one of claims 3 to 26, wherein the method comprises determining whether expression of interleukin 11 (IL-11) or a receptor for IL-11 (IL-11 R) is upregulated in the subject and administering the agent to a subject in which expression of interleukin 11 (IL-11 ) ora receptor for IL-11 (IL-11 R) is upregulated.

Description:
Treatment and Prevention of Alcoholic Liver Disease

This application claims priority from GB 2110721.4 filed 26 July 2021 and GB 2110862.6 filed 28 July 2021 , the contents and elements of which are herein incorporated by reference for all purposes.

Technical Field

The present invention relates to the diagnosis, treatment and prophylaxis of alcoholic liver disease.

Background

Alcoholic hepatitis (AH) reflects acute exacerbation of alcoholic liver disease (ALD) and is a growing healthcare burden worldwide with extremely limited treatment options.

Chronic alcohol consumption evokes physical and psychiatric disabilities and contributes to approximately 3 million deaths each year worldwide. Overall, ethanol abuse is responsible for 5.1% of the global burden of disease [Lancet (2018) 392:1015-1035] Alcoholic liver disease (ALD) is a common liver disease and is a complex pro-inflammatory process leading to steatosis, alcoholic hepatitis (AH), fibrosis, cirrhosis, and finally hepatocellular carcinoma [Asrani etal., J Hepatol (2019) 70:151-171] AH in particular reflects acute exacerbation of ALD and is a growing healthcare burden worldwide with extremely limited treatment options.

The pathogenesis of ALD, especially AH, is poorly understood which is reflected by poor therapeutic options and patient outcome. Along with direct toxicity of ethanol and acetaldehyde, alcohol causes gut dysbiosis, which subsequently provokes an altered gut barrier function and endotoxemia [Avila et al., Gut (2020) 69:764-780; Lang etal., Gut Microbes (2020) 12:1785251 ; Parlesak etal., J Hepatol (2000) 32:742-747] This dysregulation also induces overexpression of different pro-inflammatory liver cytokines which contribute to cell necrosis and liver injury [Tilg and Diehl, N Engl J Med (2000) 343:1467-1476] Interleukin (IL)-6, tumor necrosis factor-a (TNF-a), and IL-1 b drive ALD in an experimental model [Lopetuso etal., Liver. Int J Mol Sci (2018) 19; Schmidt-Arras and Rose-John, J Hepatol (2016) 64:1403- 1415; Barbier et al., Front Immunol (2019) 10:2014).

IL-11 -mediated signalling has recently been implicated in the pathology of non-alcoholic liver disease [Widjaja et al., Gastroenterology (2019) 157:777-792. e714], however the role of IL-11 -mediated signalling in alcoholic liver disease is unknown.

Summary

In a first aspect, the present disclosure provides an agent capable of inhibiting interleukin 11 (IL-11)- mediated signalling for use in a method of treating or preventing alcoholic liver disease.

Also provided is an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling in the manufacture of a medicament for use in a method of treating or preventing alcoholic liver disease. Also provided is a method of treating or preventing alcoholic liver disease, comprising administering a therapeutically or prophylactically effective amount of an agent capable of inhibiting interleukin 11 (IL-11 )- mediated signalling to a subject.

In some embodiments, the agent is an agent capable of preventing or reducing the binding of interleukin 11 (IL-11) to a receptor for interleukin 11 (IL-11 R).

In some embodiments, the agent is capable of binding to interleukin 11 (IL-11) or a receptor for interleukin 11 (IL-11 R).

In some embodiments, the agent is selected from the group consisting of: an antibody or an antigenbinding fragment thereof, a polypeptide, a peptide, a nucleic acid, an oligonucleotide, an aptamer or a small molecule.

In some embodiments, the agent is an antibody or an antigen-binding fragment thereof.

In some embodiments, the agent is an anti-IL-11 antibody antagonist of IL-11 -mediated signalling, or an antigen-binding fragment thereof.

In some embodiments, the antibody or antigen-binding fragment comprises:

(i) a heavy chain variable (VH) region incorporating the following CDRs:

HC-CDR1 having the amino acid sequence of SEQ ID NO:34 HC-CDR2 having the amino acid sequence of SEQ ID NO:35 HC-CDR3 having the amino acid sequence of SEQ ID NO:36; and

(ii) a light chain variable (VL) region incorporating the following CDRs:

LC-CDR1 having the amino acid sequence of SEQ ID NO:37 LC-CDR2 having the amino acid sequence of SEQ ID NO:38 LC-CDR3 having the amino acid sequence of SEQ ID NO:39.

In some embodiments, the antibody or antigen-binding fragment comprises:

(i) a heavy chain variable (VH) region incorporating the following CDRs:

HC-CDR1 having the amino acid sequence of SEQ ID NO:40 HC-CDR2 having the amino acid sequence of SEQ ID NO:41 HC-CDR3 having the amino acid sequence of SEQ ID NO:42; and

(ii) a light chain variable (VL) region incorporating the following CDRs:

LC-CDR1 having the amino acid sequence of SEQ ID NO:43 LC-CDR2 having the amino acid sequence of SEQ ID NO:44 LC-CDR3 having the amino acid sequence of SEQ ID NO:45.

In some embodiments, the agent is an anti-IL-11Ra antibody antagonist of IL-11 -mediated signalling, or an antigen-binding fragment thereof. In some embodiments, the antibody or antigen-binding fragment comprises:

(i) a heavy chain variable (VH) region incorporating the following CDRs:

HC-CDR1 having the amino acid sequence of SEQ ID NO:46 HC-CDR2 having the amino acid sequence of SEQ ID NO:47 HC-CDR3 having the amino acid sequence of SEQ ID NO:48; and

(ii) a light chain variable (VL) region incorporating the following CDRs:

LC-CDR1 having the amino acid sequence of SEQ ID NO:49 LC-CDR2 having the amino acid sequence of SEQ ID NO:50 LC-CDR3 having the amino acid sequence of SEQ ID NO:51.

In some embodiments, the agent is a decoy receptor.

In some embodiments, the agent is a decoy receptor for IL-11.

In some embodiments, the decoy receptor for IL-11 comprises: (i) an amino acid sequence corresponding to the cytokine binding module of gp130 and (ii) an amino acid sequence corresponding to the cytokine binding module of IL-11 Ra.

In some embodiments, the agent is an IL-11 mutein.

In some embodiments, the IL-11 mutein is W147A.

In some embodiments, the agent is capable of preventing or reducing the expression of interleukin 11 (IL- 11) or a receptor for interleukin 11 (IL-11 R).

In some embodiments, the agent is an oligonucleotide or a small molecule.

In some embodiments, the agent is an antisense oligonucleotide capable of preventing or reducing the expression of IL-11.

In some embodiments, the antisense oligonucleotide capable of preventing or reducing the expression of IL-11 is siRNA targeted to IL11 comprising the sequence of SEQ ID NO:12, 13, 14 or 15.

In some embodiments, the agent is an antisense oligonucleotide capable of preventing or reducing the expression of IL-11 Ra.

In some embodiments, the antisense oligonucleotide capable of preventing or reducing the expression of IL-11Ra is siRNA targeted to IL11RA comprising the sequence of SEQ ID NO:16, 17, 18 or 19.

In some embodiments, the interleukin 11 receptor is or comprises IL-11 Ra. In some embodiments, the method comprises administering the agent to a subject in which expression of interleukin 11 (IL-11) or a receptor for IL-11 (IL-11 R) is upregulated.

In some embodiments, the method comprises administering the agent to a subject in expression of interleukin 11 (IL-11) or a receptor for interleukin 11 (IL-11 R) has been determined to be upregulated.

In some embodiments, the method comprises determining whether expression of interleukin 11 (IL-11) or a receptor for IL-11 (IL-11 R) is upregulated in the subject and administering the agent to a subject in which expression of interleukin 11 (IL-11) or a receptor for IL-11 (IL-11 R) is upregulated.

Description

In the present disclosure the inventors establish IL-11 -mediated signalling as a driver of the pathology of alcoholic liver disease (ALD). Expression of IL-11 is found to be upregulated in the livers of mice in a model of ALD. Treatment of mice having ALD with antibody antagonist of IL-11 -mediated signalling is shown to reduce the symptoms of ALD. Thus, the present disclosure identifies the IL-11 /IL-11 receptor signalling pathway as a therapeutic target for ALD, and demonstrates that antagonism of IL-11 -mediated signalling is a suitable intervention for ALD.

Interleukin 11 and receptors for IL-11 Interleukin 11 (IL-11), also known as adipogenesis inhibitory factor, is a pleiotropic cytokine and a member of the IL-6 family of cytokines that includes IL-6, IL-11 , IL-27, IL-31, oncostatin M (OSM), leukemia inhibitory factor (LIF), cardiotrophin-1 (CT-1), cardiotrophin-like cytokine (CLC), ciliary neurotrophic factor (CNTF) and neuropoetin (NP-1).

Interleukin 11 (IL-11) is expressed in a variety of mesenchymal cell types. IL-11 genomic sequences have been mapped onto chromosome 19 and the centromeric region of chromosome 7, and is transcribed with a canonical signal peptide that ensures efficient secretion from cells. The activator protein complex of IL- 11 , cJun/AP-1 , located within its promoter sequence is critical for basal transcriptional regulation of IL-11 (Du and Williams., Blood 1997, Vol 89: 3897-3908). The immature form of human IL-11 is a 199 amino acid polypeptide whereas the mature form of IL-11 encodes a protein of 178 amino acid residues (Garbers and Scheller., Biol. Chem. 2013; 394(9): 1145-1161). The human IL-11 amino acid sequence is available under UniProt accession no. P20809 (P20809.1 GL124294; SEQ ID NO:1). Recombinant human IL-11 (oprelvekin) is also commercially available. IL-11 from other species, including mouse, rat, pig, cow, several species of bony fish and primates, have also been cloned and sequenced.

In this specification “IL-11 ” refers to an IL-11 from any species and includes isoforms, fragments, variants or homologues of an IL-11 from any species. In preferred embodiments the species is human (Homo sapiens). Isoforms, fragments, variants or homologues of an IL-11 may optionally be characterised as having at least 70%, preferably one of 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of immature or mature IL-11 from a given species, e.g. human. Isoforms, fragments, variants or homologues of an IL-11 may optionally be characterised by ability to bind IL-11 Ra (preferably from the same species) and stimulate signal transduction in cells expressing IL-11Ra and gp130 (e.g. as described in Curtis etal. Blood, 1997, 90(11); or Karpovich etal. Mol. Hum. Reprod. 20039(2): 75-80). A fragment of IL-11 may be of any length (by number of amino acids), although may optionally be at least 25% of the length of mature IL-11 and may have a maximum length of one of 50%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of the length of mature IL-11. A fragment of IL-11 may have a minimum length of 10 amino acids, and a maximum length of one of 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 or 195 amino acids.

IL-11 signals through a homodimer of the ubiquitously expressed glycoprotein 130 (gp130; also known as glycoprotein 130, IL-6ST, IL-6-beta or CD130). Gp130 is a transmembrane protein that forms one subunit of the type I cytokine receptor with the IL-6 receptor family. Specificity is gained through an individual interleukin 11 receptor subunit alpha (IL-11 Ra), which does not directly participate in signal transduction, although the initial cytokine binding event to the a-receptor leads to the final complex formation with gp130.

Human gp130 (including the 22 amino acid signal peptide) is a 918 amino acid protein, and the mature form is 866 amino acids, comprising a 597 amino acid extracellular domain, a 22 amino acid transmembrane domain, and a 277 amino acid intracellular domain. The extracellular domain of the protein comprises the cytokine-binding module (CBM) of gp130. The CBM of gp130 comprises the Ig-like domain D1, and the fibronectin-type III domains D2 and D3 of gp130. The amino acid sequence of human gp130 is available under UniProt accession no. P40189-1 (SEQ ID NO:2).

Human IL-11Ra is a 422 amino acid polypeptide (UniProt Q14626; SEQ ID NO:3) and shares ~85% nucleotide and amino acid sequence identity with the murine IL-11 Ra. Two isoforms of IL-11 Ra have been reported, which differ in the cytoplasmic domain (Du and Williams, supra). The IL-11 receptor a- chain (IL-11 Ra) shares many structural and functional similarities with the IL-6 receptor a-chain (IL-6Ra). The extracellular domain shows 24% amino acid identity including the characteristic conserved Trp-Ser- X-Trp-Ser (WSXWS) motif. The short cytoplasmic domain (34 amino acids) lacks the Box 1 and 2 regions that are required for activation of the JAK/STAT signalling pathway.

The receptor binding sites on murine IL-11 have been mapped and three sites - sites I, II and III - identified. Binding to gp130 is reduced by substitutions in the site II region and by substitutions in the site III region. Site III mutants show no detectable agonist activity and have IL-11Ra antagonist activity (Cytokine Inhibitors Chapter 8; edited by Gennaro Ciliberto and Rocco Savino, Marcel Dekker, Inc. 2001).

In this specification a receptor for IL-11 (IL-11 R) refers to a polypeptide or polypeptide complex capable of binding IL-11. In some embodiments an IL-11 receptor is capable of binding IL-11 and inducing signal transduction in cells expressing the receptor. An IL-11 receptor may be from any species and includes isoforms, fragments, variants or homologues of an IL-11 receptor from any species. In preferred embodiments the species is human (Homo sapiens).

In some embodiments the IL-11 receptor may be IL-11 Ra. In some embodiments a receptor for IL-11 may be a polypeptide complex comprising IL-11 Ra. In some embodiments the IL-11 receptor may be a polypeptide complex comprising IL-11 Ra and gp130. In some embodiments the IL-11 receptor may be gp130 or a complex comprising gp130 to which IL-11 binds.

Isoforms, fragments, variants or homologues of an IL-11Ra may optionally be characterised as having at least 70%, preferably one of 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of IL-11 Ra from a given species, e.g. human. Isoforms, fragments, variants or homologues of an IL-11Ra may optionally be characterised by ability to bind IL-11 (preferably from the same species) and stimulate signal transduction in cells expressing the IL-11Ra and gp130 (e.g. as described in Curtis etal. Blood, 1997, 90(11) or Karpovich et at. Mol. Hum. Reprod. 2003 9(2): 75-80). A fragment of an IL-11 receptor may be of any length (by number of amino acids), although may optionally be at least 25% of the length of the mature IL-11 Ra and have a maximum length of one of 50%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of the length of the mature IL-11 Ra. A fragment of an IL-11 receptor fragment may have a minimum length of 10 amino acids, and a maximum length of one of 15, 20, 25, 30, 40, 50, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 400, or 415 amino acids.

IL-11 signalling

IL-11 binds to IL-11 Ra with low affinity (Kd ~ 22 nM; see Metcalfe et al., JBC (2020) Manuscript RA119.012351), and interaction between these binding partners alone is insufficient to transduce a biological signal. The generation of a high affinity receptor (Kd -400 to 800 pmol/L) capable of signal transduction requires co-expression of the IL-11Ra and gp130 (Curtis et al Blood 1997; 90 (11):4403-12; Hilton etal., EMBO J 13:4765, 1994; Nandurkar et al., Oncogene 12:585, 1996). Binding of IL-11 to cell- surface IL-11 Ra induces heterodimerization, tyrosine phosphorylation, activation of gp130 and downstream signalling, predominantly through the mitogen-activated protein kinase (MAPK)-cascade and the Janus kinase/signal transducer and activator of transcription (Jak/STAT) pathway (Garbers and Scheller, supra).

In principle, a soluble IL-11 Ra can also form biologically active soluble complexes with IL-11 (Pflanz et al., 1999 FEBS Lett, 450, 117-122) raising the possibility that, similar to IL-6, IL-11 may in some instances bind soluble IL-11Ra prior to binding cell-surface gp130 (Garbers and Scheller, supra). Curtis et al (Blood 1997 Dec 1 ;90 (11):4403-12) describe expression of a soluble murine IL-11 receptor alpha chain (sIL- 11R) and examined signalling in cells expressing gp130. In the presence of gp130 but not transmembrane IL-11 R the slL-11 R mediated IL-11 dependent differentiation of M1 leukemic cells and proliferation in Ba/F3 cells and early intracellular events including phosphorylation of gp130, STAT3 and SHP2 similar to signalling through transmembrane IL-11 R. Activation of signalling through cell-membrane bound gp130 by IL-11 bound to soluble IL-11 Ra has recently been demonstrated (Lokau et al., 2016 Cell Reports 14, 1761-1773). This so-called IL-11 trans signalling may be important for disease pathogenesis, yet its role in human disease has not yet been studied.

As used herein, ‘IL-11 trans signalling’ is used to refer to signalling which is triggered by binding of IL-11 bound to IL-11 Ra , to gp130. The IL-11 may be bound to IL-11 Ra as a non-covalent complex. The gp130 is membrane-bound and expressed by the cell in which signalling occurs following binding of the IL-11 : 1 L- 11Ra complex to gp130. In some embodiments the IL-11Ra may be a soluble IL-11Ra. In some embodiments, the soluble IL-11 Ra is a soluble (secreted) isoform of IL-11 Ra (e.g. lacking a transmembrane domain). In some embodiments, the soluble IL-11 Ra is the liberated product of proteolytic cleavage of the extracellular domain of cell membrane bound IL-11 Ra. In some embodiments, the IL-11Ra may be cell membrane-bound, and signalling through gp130 may be triggered by binding of IL-11 bound to cell-membrane-bound IL-11 Ra, termed “IL-11 cis signalling”. In preferred embodiments, inhibition of IL-11 -mediated signalling is achieved by disrupting IL-11 -mediated cis signalling.

IL-11 -mediated signalling has been shown to stimulate hematopoiesis and thrombopoiesis, stimulate osteoclast activity, stimulate neurogenesis, inhibit adipogenesis, reduce pro inflammatory cytokine expression, modulate extracellular matrix (ECM) metabolism, and mediate normal growth control of gastrointestinal epithelial cells (Du and Williams, supra).

The physiological role of Interleukin 11 (IL-11) remains unclear. IL-11 has been most strongly linked with activation of haematopoetic cells and with platelet production. IL-11 has also been shown to confer protection against graft-vs-host-disease, inflammatory arthritis and inflammatory bowel disease, leading to IL-11 being considered an anti-inflammatory cytokine (Putoczki and Ernst, J Leukoc Biol 2010,

88(6):1109-1117). However, it is suggested that IL-11 is pro-inflammatory as well as anti-inflammatory, pro-angiogenic and important for neoplasia. Recent studies have shown that IL-11 is readily detectable during viral-induced inflammation in a mouse arthritis model and in cancers, suggesting that the expression of IL-11 can be induced by pathological stimuli. IL-11 is also linked to Stat3-dependent activation of tumour-promoting target genes in neoplastic gastrointestinal epithelium (Putoczki and Ernst, supra).

As used herein, “IL-11 signalling” and “IL-11 -mediated signalling” refers to signalling mediated by binding of IL-11 , or a fragment thereof having the function of the mature IL-11 molecule, to a receptor for IL-11. It will be appreciated that “IL-11 signalling” and “IL-11 mediated signalling” refer to signalling initiated by IL- 11 /functional fragment thereof, e.g. through binding to a receptor for IL-11. “Signalling” in turn refers to signal transduction and other cellular processes governing cellular activity.

Alcoholic liver disease

The present disclosed is concerned with the treatment and/or prevention of alcoholic liver disease (ALD).

As used herein, “alcoholic liver disease” (ALD) refers to any disease/condition associated with (e.g. caused by or characterised by) alcohol-induced perturbation to normal ( i.e . healthy, non-diseased) liver function and/or morphology. ALD may be characterised by alcohol-induced damage to the liver, alcohol- induced damage to hepatic tissue and/or alcohol-induced damage to one or more hepatic cells.

ALD encompasses alcoholic fatty liver (AFL; also known as alcoholic hepatic steatosis), alcoholic hepatitis (including e.g. alcoholic steatohepatitis (ASH)), alcohol-induced liver fibrosis, alcohol-induced cirrhosis, and alcohol-induced liver cancer (such as hepatocellular carcinoma). Accordingly, in some embodiments in accordance with the aspects and embodiments described herein, the alcoholic liver disease is selected from: alcoholic fatty liver (AFL), alcoholic hepatitis, alcoholic steatohepatitis (ASH), alcohol-induced liver fibrosis, alcohol-induced cirrhosis, and alcohol-induced liver cancer (e.g. alcohol- induced hepatocellular carcinoma). In some embodiments, the alcoholic liver disease is selected from: alcoholic fatty liver (AFL), alcoholic hepatitis and alcoholic steatohepatitis (ASH).

ALD is reviewed e.g. in Seitz etai, Nature Reviews Disease Primers (2018) 4:16, Seitz et al., J. Clin.

Med. (2021) 10: 858 and Osna etai, Alcohol Res. 2017; 38(2): 147-161, all of which are hereby incorporated by reference in its entirety. ALD arises as a consequence of (excess) alcohol consumption, and may also be referred to as alcohol-related liver disease (ARLD). Excess alcohol consumption in accordance with the present disclosure may refer to regular (e.g. 3 or more days/week) consumption of >40 g ethanol/day for a male subject, and regular consumption of >20 g ethanol/day for a female subject.

Chronic (excessive) alcohol consumption results in the accumulation of fat (mostly in the form of triglycerides, phospholipids and cholesterol esters) in hepatocytes (i.e. hepatocyte steatosis), resulting in AFL.

AFL can progress to alcoholic steatohepatitis (ASH), which is characterised by injury to hepatic tissue and liver inflammation (i.e. hepatitis). More specifically, ASH may be characterised by injury to hepatocytes (with an associated increase in serum transaminase activity), hepatocyte ballooning, the presence of Mallory-Denk bodies in hepatocytes, lobular inflammation, activated Kupffer cells, and/or infiltration of granulocytes (particularly neutrophils) into hepatic tissue.

ASH may in turn progress to alcohol-induced liver fibrosis, alcohol-induced cirrhosis, which are characterised by excess deposition of extracellular matrix components by activated, profibrotic hepatic stellate cells (HSCs). Early stage alcoholic liver fibrosis typically presents as pericellular fibrosis, which is characterised by extracellular matrix deposition along the sinusoids and around small groups of hepatocytes. Later-stage fibrosis and cirrhosis are characterised by extensive fibrosis, narrowing of hepatic vasculature (including sinusoids), portal hypertension, hepatocyte death and significant impairment of liver function (liver failure). Alcohol-induced fibrosis and cirrhosis can further give rise to hepatocellular carcinoma (HCC); indeed, cirrhosis is the most potent risk factor for the development of HCC.

A subject having alcoholic liver disease may have one or more symptoms/correlates of alcoholic liver disease. In some embodiments, a subject having alcoholic liver disease may have one or more of: hepatocyte steatosis, injury to hepatocytes, injury to hepatic tissue, hepatatis, elevated serum aspartate aminotransferase (AST), elevated serum alanine aminotransferase (ALT), hepatocyte ballooning, hepatocytes comprising Mallory-Denk bodies, lobular inflammation, activated Kupffer cells, hepatic tissue comprising granulocyte (e.g. neutrophil) infiltration, hepatic fibrosis, activated HSCs, pericellular fibrosis, cirrhosis, narrowing of hepatic vasculature, portal hypertension, hepatocyte death, liver failure, and hepatocellular carcinoma (HCC).

A subject having alcoholic liver disease may have been diagnosed as having alcoholic liver disease. A subject may satisfy the diagnostic criteria for the diagnosis of alcoholic liver disease. The diagnosis of alcoholic liver disease is describe e.g. in Torruellas etai, World J Gastroenterol. (2014) 20(33): 11684- 11699, which is hereby incorporated by reference in its entirety. ALD can generally be diagnosed based on clinical and laboratory features alone in patients with a history of significant alcohol consumption, where other etiologies for chronic liver disease have been eliminated. In general, ALD should be suspected in patients with a significant history of alcohol consumption and presenting with abnormal serum transaminases (particularly where the serum AST level is greater than the serum ALT level), hepatomegaly, clinical signs of chronic liver disease, radiographic evidence of hepatic steatosis or fibrosis/cirrhosis, or who have had a liver biopsy showing macrovesicular steatosis or cirrhosis.

Genetic risk factors for ALD have been identified by genome-wide association studies, and include genetic variants of PNPLA3 (e.g. rs738409-G, M148I), TM6SF2 (e.g. rs58542926-T, E167K), MBOAT7 (e.g. rs641738-T), MARC1 (e.g. rs2642438-C/G/T) and HNRNPUL 1 (e.g. rs15052-C). In some embodiments, a subject having alcoholic liver disease in accordance with the present disclosure may comprise one or more copies of one or more of the following alleles: PNPLA3 comprising rs738409-G, TM6SF2 comprising rs58542926-T, MBOAT7 comprising rs641738-T, MARC1 comprising rs2642438- C/G/T, and HNRNPUL1 comprising rs15052-C.

Agents capable of inhibiting the action of IL-11 Aspects of the present invention involve inhibition/antagonism of IL-11 -mediated signalling.

Herein, ‘inhibition’ refers to a reduction, decrease or lessening relative to a control condition. For example, inhibition of the action of IL-11 by an agent capable of inhibiting IL-11 -mediated signalling refers to a reduction, decrease or lessening of the extent/degree of IL-11 -mediated signalling in the absence of the agent, and/or in the presence of an appropriate control agent.

Inhibition may herein also be referred to as neutralisation or antagonism. That is, an agent capable of inhibiting IL-11 -mediated signalling (e.g. interaction, signalling or other activity mediated by IL-11 or an IL- 11 -containing complex) may be said to be a ‘neutralising’ or ‘antagonist’ agent with respect to the relevant function or process. For example, an agent which is capable of inhibiting IL-11 -mediated signalling may be referred to as an agent which is capable of neutralising IL-11 -mediated signalling, or may be referred to as an antagonist of IL-11 -mediated signalling. The IL-11 signalling pathway offers multiple routes for inhibition of IL-11 signalling. An agent capable of inhibiting IL-11 -mediated signalling may do so e.g. through inhibiting the action of one or more factors involved in, or necessary for, signalling through a receptor for IL-11.

For example, inhibition of IL-11 signalling may be achieved by disrupting interaction between IL-11 (or an IL-11 containing complex, e.g. a complex of IL-11 and IL-11 Ra) and a receptor for IL-11 (e.g. IL-11 Ra, a receptor complex comprising IL-11 Ra, gp130 or a receptor complex comprising IL-11 Ra and gp130). In some embodiments, inhibition of IL-11 -mediated signalling is achieved by inhibiting the gene or protein expression of one or more of e.g. IL-11 , IL-11 Ra and gp130.

Inhibition of IL-11 -mediated signalling may also be achieved by disrupting interaction between IL-11 :11 receptor complexes ( i.e . complexes comprising IL-11 and IL-11 Ra, or IL-11 and gp130, or IL-11 , IL-11 Ra and gp130) to form multimers (e.g. hexameric complexes) required for activation of downstream signalling by cells expressing IL-11 receptors.

In embodiments, inhibition of IL-11 -mediated signalling is achieved by disrupting IL-11 -mediated cis signalling but not disrupting IL-11 -mediated trans signalling, e.g. inhibition of IL-11 -mediated signalling is achieved by inhibiting gp130-mediated cis complexes involving membrane bound IL-11Ra. In embodiments, inhibition of IL-11 -mediated signalling is achieved by disrupting IL-11 -mediated trans signalling but not disrupting IL-11 -mediated cis signalling, i.e. inhibition of IL-11 -mediated signalling is achieved by inhibiting gp130-mediated trans signalling complexes such as IL-11 bound to soluble IL- 11Ra or IL-6 bound to soluble IL-6R. In embodiments, inhibition of IL-11 -mediated signalling is achieved by disrupting IL-11 -mediated cis signalling and IL-11 -mediated trans signalling. Any agent as described herein may be used to inhibit IL-11 -mediated cis and/or trans signalling.

In other examples, inhibition of IL-11 signalling may be achieved by disrupting signalling pathways downstream of IL-11/IL-11 Ra/gp130. That is, in some embodiments inhibition/antagonism of IL-11 - mediated signalling comprises inhibition of a signalling pathway/process/factor downstream of signalling through the IL-11/IL-11 receptor complex.

In some embodiments inhibition/antagonism of IL-11 -mediated signalling comprises inhibition of signalling through an intracellular signalling pathway which is activated by the IL-11/IL-11 receptor complex. In some embodiments inhibition/antagonism of IL-11 -mediated signalling comprises inhibition of one or more factors whose expression/activity is upregulated as a consequence of signalling through the IL- 11 /IL-11 receptor complex.

In some embodiments, the methods of the present invention employ agents capable of inhibiting JAK/STAT signalling. In some embodiments, agents capable of inhibiting JAK/STAT signalling are capable of inhibiting the action of JAK1 , JAK2, JAK3, TYK2, STAT1 , STAT2, STAT3, STAT4, STAT5A, STAT5B and/or STAT6. For example, agents may be capable of inhibiting activation of JAK/STAT proteins, inhibiting interaction of JAK or ST AT proteins with cell surface receptors e.g. IL-11 Ra or gp130, inhibiting phosphorylation of JAK proteins, inhibiting interaction between JAK and STAT proteins, inhibiting phosphorylation of STAT proteins, inhibiting dimerization of STAT proteins, inhibiting translocation of STAT proteins to the cell nucleus, inhibiting binding of STAT proteins to DNA, and/or promoting degradation of JAK and/or STAT proteins. In some embodiments, a JAK/STAT inhibitor is selected from Ruxolitinib (Jakafi/Jakavi; Incyte), Tofacitinib (Xeljanz/Jakvinus; NIH/Pfizer), Oclacitinib (Apoquel), Baricitinib (Olumiant; Incyte/Eli Lilly), Filqotinib (G-146034/GLPG-0634; Galapagos NV), Eli Lilly), Lestaurtinib (CEP-701 ; Teva), Momelotinib (GS-0387/CYT-387; Gilead Sciences), Pacritinib (SB1518; CTI), PF-04965842 (Pfizer), Upadacitinib (ABT-494; AbbVie), Peficitinib (ASP015K/JNJ-54781532; Astellas), Fedratinib (SAR302503; Celgene), Cucurbitacin I (JSI-124) and CHZ868.

In some embodiments, the methods of the present invention employ agents capable of inhibiting MAPK/ERK signalling. In some embodiments, agents capable of inhibiting MAPK/ERK signalling are capable of inhibiting the action of GRB2, inhibiting the action of RAF kinase, inhibiting the action of MEK proteins, inhibiting the activation of MAP3K/MAP2K/MAPK and/or Myc, and/or inhibiting the phosphorylation of STAT proteins. In some embodiments, agents capable of inhibiting ERK signalling are capable of inhibiting ERK p42/44. In some embodiments, an ERK inhibitor is selected from SCH772984, SC1 , VX-11e, DEL-22379, Sorafenib (Nexavar; Bayer/Onyx), SB590885, PLX4720, XL281, RAF265 (Novartis), encorafenib (LGX818/Braftovi; Array BioPharma), dabrafenib (Tafinlar; GSK), vemurafenib (Zelboraf; Roche), cobimetinib (Cotellic; Roche), CI-1040, PD0325901, Binimetinib (MEK162/MEKTOVI; Array BioPharma), selumetinib (AZD6244; Array/AstraZeneca) and Trametinib (GSK1120212/Mekinist; Novartis). In some embodiments, the methods of the present invention employ agents capable of inhibiting c-Jun N-terminal kinase (JNK) signalling/activity. In some embodiments, agents capable of inhibiting JNK signalling/activity are capable of inhibiting the action and/or phosphorylation of a JNK (e.g. JNK1 , JNK2). In some embodiments, a JNK inhibitor is selected from SP600125, CEP 1347, TCS JNK 6o, c-JUN peptide, SU3327, AEG 3482, TCS JNK 5a, BI78D3, IQ3, SR3576, IQ1S, JIP-1 (153-163) and CC401 dihydrochloride.

In the present Examples the inventors demonstrate that NOX4 expression and activity is upregulated by signalling through I L- 11 /I L- 11 Ra/g p 130. NOX4 is an NADPH oxidase, and a source of reactive oxygen species (ROS). Expression of Nox4 is upregulated in transgenic mice with hepatocyte-specific 1111 expression, and primary human hepatocytes stimulated with IL11 upregulate NOX4 expression.

In some embodiments, the present invention employs agents capable of inhibiting NOX4 expression (gene or protein expression) or function. In some embodiments, the present invention employs agents capable of inhibiting I L-11 -mediated upregulation of NOX4 expression/function. Agents capable of inhibiting NOX4 expression or function may be referred to herein as NOX4 inhibitors. For example, a NOX4 inhibitor may be capable of reducing expression (e.g. gene and/or protein expression) of NOX4, reducing the level of RNA encoding NOX4, reduce the level of NOX4 protein, and/or reducing the level of a NOX4 activity (e.g. reducing NOX4-mediated NADPH oxidase activity and/or NOX4-mediated ROS production). NOX4 inhibitors include a NOX4-binding molecules and molecules capable of reducing NOX4 expression. NOX4-binding inhibitors include peptide/nucleic acid aptamers, antibodies (and antibody fragments) and fragments of interaction partners for NOX4 which behave as antagonists of NOX4 function, and small molecules inhibitors of NOX4. Molecules capable of reducing NOX4 expression include antisense RNA (e.g. siRNA, shRNA) to NOX4. In some embodiments, a NOX4 inhibitor is selected from a NOX4 inhibitor described in Altenhofer et al., Antioxid Redox Signal. (2015) 23(5): 406-427 or Augsburder et al., Redox Biol. (2019) 26: 101272, such as GKT137831.

Binding agents

In some embodiments, agents capable of inhibiting IL-11 -mediated signalling may bind to IL-11. In some embodiments, agents capable of inhibiting IL-11 -mediated signalling may bind to a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11Ra and/or gp130). Binding of such agents may inhibit IL- 11 -mediated signalling by reducing/preventing the ability of IL-11 to bind to receptors for IL-11 , thereby inhibiting downstream signalling. Binding of such agents may inhibit IL-11 mediated cis and/or trans- signalling by reducing/preventing the ability of IL-11 to bind to receptors for IL-11 , e.g. IL-11 Ra and/or gp130, thereby inhibiting downstream signalling. Agents may bind to irans-signalling complexes such as IL-11 and soluble IL-11 Ra and inhibit gp130-mediated signalling.

Agents capable of binding to IL-11 /an IL-11 containing complex or a receptor for IL-11 may be of any kind, but in some embodiments the agent may be an antibody, an antigen-binding fragment thereof, a polypeptide, a peptide, a nucleic acid, an oligonucleotide, an aptamer or a small molecule. The agents may be provided in isolated or purified form, or may be formulated as a pharmaceutical composition or medicament.

Antibodies and antigen-binding fragments

In some embodiments, an agent capable of binding to IL-11/an IL-11 containing complex or a receptor for IL-11 is an antibody, or an antigen-binding fragment thereof. In some embodiments, an agent capable of binding to IL-11/an IL-11 containing complex or a receptor for IL-11 is a polypeptide, e.g. a decoy receptor molecule. In some embodiments, an agent capable of binding to IL-11/an IL-11 containing complex or a receptor for IL-11 may be an aptamer.

In some embodiments, an agent capable of binding to IL-11/an IL-11 containing complex or a receptor for IL-11 is an antibody, or an antigen-binding fragment thereof. An “antibody” is used herein in the broadest sense, and encompasses monoclonal antibodies, polyclonal antibodies, monospecific and multispecific antibodies (e.g., bispecific antibodies), and antibody fragments, as long as they display binding to the relevant target molecule.

In view of today's techniques in relation to monoclonal antibody technology, antibodies can be prepared to most antigens. The antigen-binding portion may be a part of an antibody (for example a Fab fragment) or a synthetic antibody fragment (for example a single chain Fv fragment [ScFv]). Monoclonal antibodies to selected antigens may be prepared by known techniques, for example those disclosed in "Monoclonal Antibodies: A manual of techniques ", H Zola (CRC Press, 1988) and in "Monoclonal Hybridoma Antibodies: Techniques and Applications ", J G R Hurrell (CRC Press, 1982). Chimaeric antibodies are discussed by Neuberger et al (1988, 8th International Biotechnology Symposium Part 2, 792-799). Monoclonal antibodies (mAbs) are particularly useful in the methods of the invention, and are a homogenous population of antibodies specifically targeting a single epitope on an antigen.

Polyclonal antibodies are also useful in the methods of the invention. Monospecific polyclonal antibodies are preferred. Suitable polyclonal antibodies can be prepared using methods well known in the art.

Antigen-binding fragments of antibodies, such as Fab and Fab2 fragments may also be used/provided as can genetically engineered antibodies and antibody fragments. The variable heavy (VH) and variable light (VL) domains of the antibody are involved in antigen recognition, a fact first recognised by early protease digestion experiments. Further confirmation was found by "humanisation" of rodent antibodies. Variable domains of rodent origin may be fused to constant domains of human origin such that the resultant antibody retains the antigenic specificity of the rodent parented antibody (Morrison et al (1984) Proc. Natl. Acad. Sd. USA 81 , 6851-6855).

Antibodies and antigen-binding fragments according to the present disclosure comprise the complementarity-determining regions (CDRs) of an antibody which is capable of binding to the relevant target molecule (i.e. IL-11/an IL-11 containing complex/a receptor for IL-11).

Antibodies capable of binding to IL-11 include e.g. monoclonal mouse anti-human IL-11 antibody clone #22626; Catalog No. MAB218 (R&D Systems, MN, USA), used e.g. in Bockhorn etal. Nat. Commun. (2013) 4(0):1393, clone 6D9A (Abbiotec), clone KT8 (Abbiotec), clone M3103F11 (BioLegend), clone 1F1 (Abnova Corporation), clone 3C6 (Abnova Corporation), clone GF1 (LifeSpan Biosciences), clone 13455 (Source BioScience), 11 h3/19.6.1 (Hermann etal., Arthritis Rheum. (1998) 41 (8):1388-97), AB-218-NA (R&D Systems), X203 (Ng etal., Sci Transl Med. (2019) 11(511) pii: eaaw1237) and anti-IL-11 antibodies disclosed in US 2009/0202533 A1 , WO 99/59608 A2, WO 2018/109174 A2 and WO 2019/238882 A1.

In particular, anti-IL-11 antibody clone 22626 (also known as MAB218) has been shown to be an antagonist of IL-11 mediated signalling, e.g. in Schaefer et al., Nature (2017) 552(7683): 110-115. Monoclonal antibody 11 h3/19.6.1 is disclosed in Hermann etal., Arthritis Rheum. (1998) 41 (8):1388-97 to be a neutralising anti-IL-11 lgG1. AB-218-NA from R&D Systems, used e.g. in McCoy etal., BMC Cancer (2013) 13:16, is another example of neutralizing anti-IL-11 antibody. WO 2018/109174 A2 and WO 2019/238882 A1 disclose yet further exemplary anti-IL-11 antibody antagonists of IL-11 mediated signalling. X203 (also referred to as Enx203) disclosed in Ng, et al., “IL-11 is a therapeutic target in idiopathic pulmonary fibrosis.” bioRxiv 336537; doi: https://doi.org/10.1101/336537 and WO 2019/238882 A1 is an anti-IL-11 antibody antagonist of IL-11 -mediated signalling, and comprises the VH region according to SEQ ID NO:92 of WO 2019/238882 A1 (SEQ ID NO:22 of the present disclosure), and the VL region according to SEQ ID NO:94 of WO 2019/238882 A1 (SEQ ID NO:23 of the present disclosure). Humanised versions of the X203 are described in WO 2019/238882 A1, including hEnx203 which comprises the VH region according to SEQ ID NO:117 of WO 2019/238882 A1 (SEQ ID NO:30 of the present disclosure), and the VL region according to SEQ ID NO:122 of WO 2019/238882 A1 (SEQ ID NO:31 of the present disclosure). Enx108A is a further example of an anti-IL-11 antibody antagonist of IL- 11 -mediated signalling, and comprises the VH region according to SEQ ID NO:8 of WO 2019/238882 A1 (SEQ ID NO:26 of the present disclosure), and the VL region according to SEQ ID NO:20 of WO 2019/238882 A1 (SEQ ID NO:27 of the present disclosure).

Antibodies capable of binding to IL-11Ra include e.g. monoclonal antibody clone 025 (Sino Biological), clone EPR5446 (Abeam), clone 473143 (R & D Systems), clones 8E2, 8D10 and 8E4 and the affinity- matured variants of 8E2 described in US 2014/0219919 A1 , the monoclonal antibodies described in Blanc et al ( J . Immunol Methods. 2000 Jul 31 ;241 (1-2);43-59), X209 (Widjaja et al., Gastroenterology (2019) 157(3)777-792, which is also published as Widjaja, et al., “IL-11 neutralising therapies target hepatic stellate cell-induced liver inflammation and fibrosis in NASH.” bioRxiv 470062; doi: https://doi.org/10.1101/470062) antibodies disclosed in WO 2014121325 A1 and US 2013/0302277 A1, and anti-IL-11Ra antibodies disclosed in US 2009/0202533 A1 , WO 99/59608 A2, WO 2018/109170 A2 and WO 2019/238884 A1.

In particular, anti-IL-11Ra antibody clone 473143 (also known as MAB1977) has been shown to be an antagonist of IL-11 mediated signalling, e.g. in Schaefer et al., Nature (2017) 552(7683): 110-115. US 2014/0219919 A1 provides sequences for anti-human IL-11Ra antibody clones 8E2, 8D10 and 8E4, and discloses their ability to antagonise IL-11 mediated signalling - see e.g. [0489] to [0490] of US 2014/0219919 A1. US 2014/0219919 A1 moreover provides sequence information for an additional 62 affinity- matured variants of clone 8E2, 61 of which are disclosed to antagonise IL-11 mediated signalling - see Table 3 of US 2014/0219919 A1. WO 2018/109170 A2 and WO 2019/238884 A1 disclose yet further exemplary anti-IL-11 Ra antibody antagonists of IL-11 mediated signalling. X209 (also referred to as Enx209) disclosed in Widjaja, et al., “IL-11 neutralising therapies target hepatic stellate cell-induced liver inflammation and fibrosis in NASH.” bioRxiv 470062; doi: https://doi.org/10.1101/470062 and WO 2019/238884 A1 is an anti-IL-11 Ra antibody antagonist of IL-11 -mediated signalling, and comprises the VH region according to SEQ ID NO:7 of WO 2019/238884 A1 (SEQ ID NO:24 of the present disclosure), and the VL region according to SEQ ID NO:14 of WO 2019/238884 A1 (SEQ ID NO:25 of the present disclosure). Humanised versions of the X209 are described in WO 2019/238884 A1, including hEnx209 which comprises the VH region according to SEQ ID NO:11 of WO 2019/238884 A1 (SEQ ID NO:32 of the present disclosure), and the VL region according to SEQ ID NO:17 of WO 2019/238884 A1 (SEQ ID NO:33 of the present disclosure).

The skilled person is well aware of techniques for producing antibodies suitable for therapeutic use in a given species/subject. For example, procedures for producing antibodies suitable for therapeutic use in humans are described in Park and Smolen Advances in Protein Chemistry (2001) 56: 369-421 (hereby incorporated by reference in its entirety). Antibodies to a given target protein (e.g. IL-11 or IL-11Ra) can be raised in model species (e.g. rodents, lagomorphs), and subsequently engineered in order to improve their suitability for therapeutic use in a given species/subject. For example, one or more amino acids of monoclonal antibodies raised by immunisation of model species can be substituted to arrive at an antibody sequence which is more similar to human germline immunoglobulin sequences (thereby reducing the potential for anti-xenogenic antibody immune responses in the human subject treated with the antibody). Modifications in the antibody variable domains may focus on the framework regions in order to preserve the antibody paratope. Antibody humanisation is a matter of routine practice in the art of antibody technology, and is reviewed e.g. in Almagro and Fransson, Frontiers in Bioscience (2008) 13:1619-1633, Safdari etai, Biotechnology and Genetic Engineering Reviews (2013) 29(2): 175-186 and Lo et al., Microbiology Spectrum (2014) 2(1), all of which are hereby incorporated by reference in their entirety. The requirement for humanisation can be circumvented by raising antibodies to a given target protein (e.g. IL-11 or IL-11 Ra) in transgenic model species expressing human immunoglobulin genes, such that the antibodies raised in such animals are fully-human (described e.g. in Bmggemann et al., Arch Immunol Ther Exp (Warsz) (2015) 63(2): 101—108, which is hereby incorporated by reference in its entirety).

Phage display techniques may also be employed to the identification of antibodies to a given target protein (e.g. IL-11 or IL-11 Ra), and are well known to the skilled person. The use of phage display for the identification of fully human antibodies to human target proteins is reviewed e.g. in Hoogenboom, Nat. Biotechnol. (2005) 23, 1105-1116 and Chan etai, International Immunology (2014) 26(12): 649-657, which are hereby incorporated by reference in their entirety.

The antibodies/fragments may be antagonist antibodies/fragments that inhibit or reduce a biological activity of IL-11. The antibodies/fragments may be neutralising antibodies that neutralise the biological effect of IL-11 , e.g. its ability to stimulate productive signalling via an IL-11 receptor. Neutralising activity may be measured by ability to neutralise IL-11 induced proliferation in the T11 mouse plasmacytoma cell line (Nordan, R. P. etai. (1987) J. Immunol. 139:813).

IL-11- or IL-11Ra-binding antibodies can be evaluated for the ability to antagonise IL-11 -mediated signalling, e.g. using the assay described in US 2014/0219919 A1 or Blanc et al (J. Immunol Methods. 2000 Jul 31;241(1-2);43-59. Briefly, IL-11- and IL-11Ra-binding antibodies can be evaluated in vitro for the ability to inhibit proliferation of Ba/F3 cells expressing IL-11Ra and gp130 from the appropriate species, in response to stimulation with IL-11 from the appropriate species. Alternatively, IL-11- and IL- 11 Ra-binding antibodies can be analysed in vitro for the ability to inhibit the fibroblast-to-myofibroblast transition following stimulation of fibroblasts with TGFpl , by evaluation of aSMA expression (as described e.g. in WO 2018/109174 A2 (Example 6) and WO 2018/109170 A2 (Example 6), Ng etai, Sci Transl Med. (2019) 11(511) pii: eaaw1237 and Widjaja etai, Gastroenterology (2019) 157(3):777-792).

Antibodies generally comprise six CDRs; three in the light chain variable region (VL): LC-CDR1 , LC- CDR2, LC-CDR3, and three in the heavy chain variable region (VH): HC-CDR1, HC-CDR2 and HC- CDR3. The six CDRs together define the paratope of the antibody, which is the part of the antibody which binds to the target molecule. The VH region and VL region comprise framework regions (FRs) either side of each CDR, which provide a scaffold for the CDRs. From N-terminus to C-terminus, VH regions comprise the following structure: N term-[HC-FR1]-[HC-CDR1]-[HC-FR2]-[HC-CDR2]-[HC-FR3]-[HC- CDR3]-[HC-FR4]-C term; and VL regions comprise the following structure: N term-[LC-FR1]-[LC-CDR1]- [LC-FR2]-[LC-CDR2]-[LC-FR3]-[LC-CDR3]-[LC-FR4]-C term.

There are several different conventions for defining antibody CDRs and FRs, such as those described in Kabat etai, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991), Chothia etai, J. Mol. Biol. 196:901-917 (1987), and VBASE2, as described in Retter et ai, Nucl. Acids Res. (2005) 33 (suppl 1): D671-D674. The CDRs and FRs of the VH regions and VL regions of the antibodies described herein are defined according to the Kabat system.

In some embodiments an antibody, or an antigen-binding fragment thereof, according to the present disclosure is derived from an antibody which binds specifically to IL-11 (e.g. Enx108A, Enx203 or hEnx203). In some embodiments an antibody, or an antigen-binding fragment thereof, according to the present disclosure is derived from an antibody which binds specifically to IL-11 Ra (e.g. Enx209 or hEnx209).

Antibodies and antigen-binding fragments according to the present disclosure preferably inhibit IL-11 - mediated signalling. Such antibodies/antigen-binding fragments may be described as being antagonists of IL-11 -mediated signalling, and/or may be described as having the ability to neutralise IL-11 -mediated signalling.

In some embodiments, the antibody/antigen-binding fragment comprises the CDRs of an antibody which binds to IL-11. In some embodiments the antibody/antigen-binding fragment comprises the CDRs of, or CDRs derived from, the CDRs of an IL-11 -binding antibody described herein (e.g. Enx108A, Enx203 or hEnx203).

In some embodiments the antibody/antigen-binding fragment comprises a VH region incorporating the following CDRs:

(1)

HC-CDR1 having the amino acid sequence of SEQ ID NO:34 HC-CDR2 having the amino acid sequence of SEQ ID NO:35 HC-CDR3 having the amino acid sequence of SEQ ID NO:36, or a variant thereof in which one or two or three amino acids in one or more of HC-CDR1 , HC- CDR2, or HC-CDR3 are substituted with another amino acid.

In some embodiments the antibody/antigen-binding fragment comprises a VL region incorporating the following CDRs:

(2)

LC-CDR1 having the amino acid sequence of SEQ ID NO:37 LC-CDR2 having the amino acid sequence of SEQ ID NO:38 LC-CDR3 having the amino acid sequence of SEQ ID NO:39, or a variant thereof in which one or two or three amino acids in one or more of LC-CDR1 , LC- CDR2, or LC-CDR3 are substituted with another amino acid.

In some embodiments the antibody/antigen-binding fragment comprises a VH region incorporating the following CDRs:

(3)

HC-CDR1 having the amino acid sequence of SEQ ID NO:40 HC-CDR2 having the amino acid sequence of SEQ ID NO:41 HC-CDR3 having the amino acid sequence of SEQ ID NO:42, or a variant thereof in which one or two or three amino acids in one or more of HC-CDR1 , HC- CDR2, or HC-CDR3 are substituted with another amino acid.

In some embodiments the antibody/antigen-binding fragment comprises a VL region incorporating the following CDRs:

(4)

LC-CDR1 having the amino acid sequence of SEQ ID NO:43 LC-CDR2 having the amino acid sequence of SEQ ID NO:44 LC-CDR3 having the amino acid sequence of SEQ ID NO:45, or a variant thereof in which one or two or three amino acids in one or more of LC-CDR1 , LC- CDR2, or LC-CDR3 are substituted with another amino acid.

In some embodiments the antibody/antigen-binding fragment comprises a VH region incorporating the CDRs according to (1), and a VL region incorporating the CDRs according to (2). In some embodiments the antibody/antigen-binding fragment comprises a VH region incorporating the CDRs according to (3), and a VL region incorporating the CDRs according to (4).

In some embodiments the antibody/antigen-binding fragment comprises the VH region and the VL region of an antibody which binds to IL-11. In some embodiments the antibody/antigen-binding fragment comprises the VH region and VL region of, or a VH region and VL region derived from, the VH region and VL region of an IL-11 -binding antibody described herein (e.g. Enx108A, Enx203 or hEnx203).

In some embodiments the antibody/antigen-binding fragment comprises a VH region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:26. In some embodiments the antibody/antigen-binding fragment comprises a VL region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:27. In some embodiments the antibody/antigen-binding fragment comprises a VH region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,

98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:26 and a VL region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:27.

In some embodiments the antibody/antigen-binding fragment comprises a VH region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:22. In some embodiments the antibody/antigen-binding fragment comprises a VL region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:23. In some embodiments the antibody/antigen-binding fragment comprises a VH region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,

98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:22 and a VL region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:23.

In some embodiments the antibody/antigen-binding fragment comprises a VH region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:30. In some embodiments the antibody/antigen-binding fragment comprises a VL region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:31. In some embodiments the antibody/antigen-binding fragment comprises a VH region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,

98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:30 and a VL region comprising an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:31.

In some embodiments, the antibody/antigen-binding fragment comprises the CDRs of an antibody which binds to IL-11 Ra. In some embodiments the antibody/antigen-binding fragment comprises the CDRs of, or CDRs derived from, the CDRs of an IL-11 Ra-binding antibody described herein (e.g. Enx209 or hEnx209). In some embodiments the antibody/antigen-binding fragment comprises a VH region incorporating the following CDRs:

(5)

HC-CDR1 having the amino acid sequence of SEQ ID NO:46 HC-CDR2 having the amino acid sequence of SEQ ID NO:47 HC-CDR3 having the amino acid sequence of SEQ ID NO:48, or a variant thereof in which one or two or three amino acids in one or more of HC-CDR1 , HC- CDR2, or HC-CDR3 are substituted with another amino acid.

In some embodiments the antibody/antigen-binding fragment comprises a VL region incorporating the following CDRs:

(6)

LC-CDR1 having the amino acid sequence of SEQ ID NO:49 LC-CDR2 having the amino acid sequence of SEQ ID NO:50 LC-CDR3 having the amino acid sequence of SEQ ID NO:51, or a variant thereof in which one or two or three amino acids in one or more of LC-CDR1 , LC- CDR2, or LC-CDR3 are substituted with another amino acid.

In some embodiments the antibody/antigen-binding fragment comprises a VH region incorporating the CDRs according to (5), and a VL region incorporating the CDRs according to (6).

In some embodiments the antibody/antigen-binding fragment comprises the VH region and the VL region of an antibody which binds to IL-11 Ra. In some embodiments the antibody/antigen-binding fragment comprises the VH region and VL region of, or a VH region and VL region derived from, the VH region and VL region of an IL-11Ra-binding antibody described herein (e.g. Enx209 or hEnx209).

In embodiments in accordance with the present invention in which one or more amino acids of a reference amino acid sequence (e.g. a CDR sequence, VH region sequence or VL region sequence described herein) are substituted with another amino acid, the substitutions may conservative substitutions, for example according to the following Table. In some embodiments, amino acids in the same block in the middle column are substituted. In some embodiments, amino acids in the same line in the rightmost column are substituted: In some embodiments, substitution(s) may be functionally conservative. That is, in some embodiments the substitution may not affect (or may not substantially affect) one or more functional properties ( e.g . target binding) of the antibody/fragment comprising the substitution relative to the equivalent unsubstituted molecule.

In some embodiments, substitution(s) relative to a reference VH region or VL region sequence may be focussed in a particular region or regions of the VH region or VL region sequence. For example, variation from a reference VH region or VL region sequence may be focussed in one or more of the framework regions (FR1 , FR2, FR3 and/or FR4).

Antibodies and antigen-binding fragments according to the present disclosure may be designed and prepared using the sequences of monoclonal antibodies (mAbs) capable of binding to the relevant target molecule. Antigen-binding regions of antibodies, such as single chain variable fragment (scFv), Fab and Fab2 fragments may also be used/provided. An ‘antigen-binding region’ or ‘antigen binding fragment’ is any fragment of an antibody which is capable of binding to the target for which the given antibody is specific.

In some embodiments the antibodies/fragments comprise the VL and VH regions of an antibody which is capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11. The VL and VH region of an antigen-binding region of an antibody together constitute the Fv region. In some embodiments the antibodies/fragments comprise or consist of the Fv region of an antibody which is capable of binding to IL- 11 , an IL-11 containing complex, or a receptor for IL-11. The Fv region may be expressed as a single chain wherein the VH and VL regions are covalently linked, e.g. by a flexible oligopeptide. Accordingly, antibodies/fragments may comprise or consist of an scFv comprising the VL and VH regions of an antibody which is capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11.

The VL and light chain constant (CL) region, and the VH region and heavy chain constant 1 (CH1) region of an antigen-binding region of an antibody together constitute the Fab region. In some embodiments the antibodies/fragments comprise or consist of the Fab region of an antibody which is capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11.

In some embodiments, antibodies/fragments comprise, or consist of, whole antibody capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11. A “whole antibody” refers to an antibody having a structure which is substantially similar to the structure of an immunoglobulin (Ig). Different kinds of immunoglobulins and their structures are described e.g. in Schroeder and Cavacini J Allergy Clin Immunol. (2010) 125(202): S41-S52, which is hereby incorporated by reference in its entirety. Immunoglobulins of type G (i.e. IgG) are ~150 kDa glycoproteins comprising two heavy chains and two light chains. From N- to C-terminus, the heavy chains comprise a VH followed by a heavy chain constant region comprising three constant domains (CH1 , CH2, and CH3), and similarly the light chain comprises a VL followed by a CL. Depending on the heavy chain, immunoglobulins may be classed as IgG (e.g. lgG1, lgG2, lgG3, lgG4), IgA ( e.g . lgA1, lgA2), IgD, IgE, or IgM. The light chain may be kappa (K) or lambda (l).

In some embodiments the antibody/antigen-binding fragment of the present disclosure comprises an immunoglobulin heavy chain constant sequence. In some embodiments, an immunoglobulin heavy chain constant sequence may be a human immunoglobulin heavy chain constant sequence. In some embodiments the immunoglobulin heavy chain constant sequence is, or is derived from, the heavy chain constant sequence of an IgG (e.g. lgG1 , lgG2, lgG3, lgG4), IgA (e.g. lgA1 , lgA2), IgD, IgE or IgM, e.g. a human IgG (e.g. hlgG1 , hlgG2, hlgG3, hlgG4), hlgA (e.g. hlgA1 , hlgA2), hlgD, hlgE or hlgM. In some the immunoglobulin heavy chain constant sequence is, or is derived from, the heavy chain constant sequence of a human lgG1 allotype (e.g. G1m1, G1m2, G1m3 orG1m17).

In some embodiments the immunoglobulin heavy chain constant sequence is, or is derived from, the constant region sequence of human immunoglobulin G 1 constant (IGHG1; UniProt: P01857-1, v1). In some embodiments the immunoglobulin heavy chain constant sequence is, or is derived from, the constant region sequence of human immunoglobulin G 1 constant (IGHG1; UniProt: P01857-1, v1) comprising substitutions K214R, D356E and L358M (i.e. the G1m3 allotype). In some embodiments the antibody/antigen-binding fragment comprises an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:52.

In some embodiments the immunoglobulin heavy chain constant sequence is, or is derived from, the constant region sequence of human immunoglobulin G 4 constant (IGHG4; UniProt: P01861, v1). In some embodiments the immunoglobulin heavy chain constant sequence is, or is derived from, the constant region sequence of human immunoglobulin G 4 constant (IGHG4; UniProt: P01861, v1) comprising substitutions S241P and/or L248E. The S241P mutation is hinge stabilising while the L248E mutation further reduces the already low ADCC effector function of lgG4 (Davies and Sutton, Immunol Rev. 2015 Nov; 268(1):139-159; Angal et al Mol Immunol. 1993 Jan;30(1):105-8). In some embodiments the antibody/antigen-binding fragment comprises an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:53.

In some embodiments the antibody/antigen-binding fragment of the present disclosure comprises an immunoglobulin light chain constant sequence. In some embodiments, an immunoglobulin light chain constant sequence may be a human immunoglobulin light chain constant sequence. In some embodiments the immunoglobulin light chain constant sequence is, or is derived from, a kappa (K) or lambda (l) light chain, e.g. human immunoglobulin kappa constant (IGKC; CK; UniProt: P01834-1 , v2), or human immunoglobulin lambda constant (IGLC; CK), e.g. IGLC1 (UniProt: P0CG04-1, v1), IGLC2 (UniProt: PODOY2-1 , v1), IGLC3 (UniProt: PODOY3-1 , v1), IGLC6 (UniProt: P0CF74-1, v1) or IGLC7 (UniProt: A0M8Q6-1 , v3).

In some embodiments the antibody/antigen-binding fragment comprises an immunoglobulin light chain constant sequence. In some embodiments the immunoglobulin light chain constant sequence is, or is derived from human immunoglobulin kappa constant (IGKC; CK; UniProt: P01834-1, v2; SEQ ID NO:90). In some embodiments the immunoglobulin light chain constant sequence is a human immunoglobulin lambda constant (IGLC; CK), e.g. IGLC1 , IGLC2, IGLC3, IGLC6 or IGLC7. In some embodiments the antibody/antigen-binding fragment comprises an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:54. In some embodiments the antibody/antigen-binding fragment comprises an amino acid sequence having at least 70% sequence identity more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the amino acid sequence of SEQ ID NO:55.

In some embodiments, the antibody/antigen-binding fragment comprises: (i) a polypeptide comprising or consisting of an amino acid sequence having at least 70%, preferably one of 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO:28, and (ii) a polypeptide comprising or consisting of an amino acid sequence having at least 70%, preferably one of 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO:29.

In some embodiments, the antibody/antigen-binding fragment comprises: (i) a polypeptide comprising or consisting of an amino acid sequence having at least 70%, preferably one of 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO:56, and (ii) a polypeptide comprising or consisting of an amino acid sequence having at least 70%, preferably one of 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO:57.

In some embodiments, the antibody/antigen-binding fragment comprises: (i) a polypeptide comprising or consisting of an amino acid sequence having at least 70%, preferably one of 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO:58, and (ii) a polypeptide comprising or consisting of an amino acid sequence having at least 70%, preferably one of 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to the amino acid sequence of SEQ ID NO:59.

Fab, Fv, ScFv and dAb antibody fragments can all be expressed in and secreted from E. coli, thus allowing the facile production of large amounts of the said fragments. Whole antibodies, and F(ab')2 fragments are "bivalent". By "bivalent" we mean that the said antibodies and F(ab')2 fragments have two antigen combining sites. In contrast, Fab, Fv, ScFvand dAb fragments are monovalent, having only one antigen combining site. Synthetic antibodies capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11 may also be made using phage display technology as is well known in the art.

Antibodies may be produced by a process of affinity maturation in which a modified antibody is generated that has an improvement in the affinity of the antibody for antigen, compared to an unmodified parent antibody. Affinity-matured antibodies may be produced by procedures known in the art, e.g., Marks etal., Rio/Technology 10:779-783 (1992); Barbas et al. Proc Nat. Acad. Sci. USA 91 :3809-3813 (1994); Schier etal. Gene 169:147-155 (1995); Yelton etal. J. Immunol. 155:1994-2004 (1995); Jackson etal., J. Immunol. 154(7):331 0-159 (1995); and Hawkins et al, J. Mol. Biol. 226:889-896 (1992).

Antibodies/fragments include bi-specific antibodies, e.g. composed of two different fragments of two different antibodies, such that the bi-specific antibody binds two types of antigen. The bispecific antibody comprises an antibody/fragment as described herein capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11. The antibody may contain a different fragment having affinity for a second antigen, which may be any desired antigen. Techniques for the preparation of bi-specific antibodies are well known in the art, e.g. see Mueller, D et al., (2010 Biodrugs 24 (2): 89-98), Wozniak- Knopp G et al. , (2010 Protein Eng Des 23 (4): 289-297), and Baeuerle, PA et al. , (2009 Cancer Res 69 (12): 4941-4944). Bispecific antibodies and bispecific antigen-binding fragments may be provided in any suitable format, such as those formats described in Kontermann MAbs 2012, 4(2): 182-197, which is hereby incorporated by reference in its entirety. For example, a bispecific antibody or bispecific antigenbinding fragment may be a bispecific antibody conjugate (e.g. an lgG2, F(ab’)2 or CovX-Body), a bispecific IgG or IgG-like molecule (e.g. an IgG, scFv4-lg, IgG-scFv, scFv-lgG, DVD-lg, IgG-sVD, sVD- IgG, 2 in 1 -IgG, mAb2, orTandemab common LC), an asymmetric bispecific IgG or IgG-like molecule (e.g. a kih IgG, kih IgG common LC, CrossMab, kih IgG-scFab, mAb-Fv, charge pair or SEED-body), a small bispecific antibody molecule (e.g. a Diabody (Db), dsDb, DART, scDb, tandAbs, tandem scFv (taFv), tandem dAb/VHH, triple body, triple head, Fab-scFv, or F(ab’)2-scFv2), a bispecific Fc and CH3 fusion protein (e.g. a taFv-Fc, Di-diabody, scDb-CH3, scFv-Fc-scFv, HCAb-VHH, scFv-kih-Fc, orscFv- kih-CH3), ora bispecific fusion protein (e.g. a scFv2-albumin, scDb-albumin, taFv-toxin, DNL-Fab3, DNL- Fab4-lgG, DNL-Fab4-lgG-cytokine2). See in particular Figure 2 of Kontermann MAbs 2012, 4(2): 182-19.

Methods for producing bispecific antibodies include chemically crosslinking antibodies or antibody fragments, e.g. with reducible disulphide or non-reducible thioether bonds, for example as described in Segal and Bast, 2001. Production of Bispecific Antibodies. Current Protocols in Immunology. 14:IV:2.13:2.13.1 —2.13.16, which is hereby incorporated by reference in its entirety. For example, N- succinimidyl-3-(-2-pyridyldithio)-propionate (SPDP) can be used to chemically crosslink e.g. Fab fragments via hinge region SH- groups, to create disulfide-linked bispecific F(ab)2 heterodimers. Other methods for producing bispecific antibodies include fusing antibody-producing hybridomas e.g. with polyethylene glycol, to produce a quadroma cell capable of secreting bispecific antibody, for example as described in D. M. and Bast, B. J. 2001. Production of Bispecific Antibodies. Current Protocols in Immunology. 14:IV:2.13:2.13.1 -2.13.16.

Bispecific antibodies and bispecific antigen-binding fragments can also be produced recombinantly, by expression from e.g. a nucleic acid construct encoding polypeptides for the antigen binding molecules, for example as described in Antibody Engineering: Methods and Protocols, Second Edition (Humana Press, 2012), at Chapter 40: Production of Bispecific Antibodies: Diabodies and Tandem scFv (Hornig and Farber-Schwarz), or French, How to make bispecific antibodies, Methods Mol. Med. 2000; 40:333-339.

For example, a DNA construct encoding the light and heavy chain variable domains for the two antigen binding domains (i.e. the light and heavy chain variable domains for the antigen binding domain capable of binding to IL-11 , an IL-11 containing complex, or a receptor for IL-11 , and the light and heavy chain variable domains for the antigen binding domain capable of binding to another target protein), and including sequences encoding a suitable linker or dimerization domain between the antigen binding domains can be prepared by molecular cloning techniques. Recombinant bispecific antibody can thereafter be produced by expression (e.g. in vitro) of the construct in a suitable host cell (e.g. a mammalian host cell), and expressed recombinant bispecific antibody can then optionally be purified.

Decoy receptors

Peptide or polypeptide based agents capable of binding to IL-11 or IL-11 containing complexes may be based on the IL-11 receptor, e.g. an IL-11 binding fragment of an IL-11 receptor.

In some embodiments, the binding agent may comprise an IL-11 -binding fragment of the IL-11Ra chain, and may preferably be soluble and/or exclude one or more, or all, of the transmembrane domain(s). In some embodiments, the binding agent may comprise an IL-11 -binding fragment of gp130, and may preferably be soluble and/or exclude one or more, or all, of the transmembrane domain(s). Such molecules may be described as decoy receptors. Binding of such agents may inhibit IL-11 mediated cis and/or irans-signalling by reducing/preventing the ability of IL-11 to bind to receptors for IL-11 , e.g. IL- 11 Ra or gp130, thereby inhibiting downstream signalling.

Curtis et al (Blood 1997 Dec 1 ;90 (11):4403-12) report that a soluble murine IL-11 receptor alpha chain (slL-11 R) was capable of antagonizing the activity of IL-11 when tested on cells expressing the transmembrane IL-11 R and gp130. They proposed that the observed IL-11 antagonism by the slL-11 R depends on limiting numbers of gp130 molecules on cells already expressing the transmembrane IL-11 R.

The use of soluble decoy receptors as the basis for inhibition of signal transduction and therapeutic intervention has also been reported for other signalling molecule:receptor pairs, e.g. VEGF and the VEGF receptor (De-Chao Yu etal., Molecular Therapy (2012); 205, 938-947; Konnerand Dupont Clin Colorectal Cancer 2004 Oct;4 Suppl 2:S81-5). As such, in some embodiments a binding agent may be a decoy receptor, e.g. a soluble receptor for IL-11 and/or IL-11 containing complexes. Competition for IL-11 and/or IL-11 containing complexes provided by a decoy receptor has been reported to lead to IL-11 antagonist action (Curtis et al., supra). Decoy IL-11 receptors are also described in WO 2017/103108 A1 and WO 2018/109168 A1 , which are hereby incorporated by reference in their entirety.

Decoy IL-11 receptors preferably bind IL-11 and/or IL-11 containing complexes, and thereby make these species unavailable for binding to gp130, IL-11 Ra and/or gp130:IL-11 Ra receptors. As such, they act as ‘decoy’ receptors for IL-11 and IL-11 containing complexes, much in the same way that etanercept acts as a decoy receptor for TNFa. IL-11 -mediated signalling is reduced as compared to the level of signalling in the absence of the decoy receptor.

Decoy IL-11 receptors preferably bind to IL-11 through one or more cytokine binding modules (CBMs). The CBMs are, or are derived from or homologous to, the CBMs of naturally occurring receptor molecules for IL-11 . For example, decoy IL-11 receptors may comprise, or consist of, one or more CBMs which are from, are derived from or homologous to the CBM of gp130 and/or IL-11Ra.

In some embodiments, a decoy IL-11 receptor may comprise, or consist of, an amino acid sequence corresponding to the cytokine binding module of gp130. In some embodiments, a decoy IL-11 receptor may comprise an amino acid sequence corresponding to the cytokine binding module of IL-11 Ra. Herein, an amino acid sequence which ‘corresponds’ to a reference region or sequence of a given peptide/polypeptide has at least 60%, e.g. one of at least 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to the amino acid sequence of the reference region/sequence.

In some embodiments a decoy receptor may be able to bind IL-11 , e.g. with binding affinity of at least 100pM or less, optionally one of 10pM or less, 1 pM or less, 10OnM or less, or about 1 to 10OnM. In some embodiments a decoy receptor may comprise all or part of the IL-11 binding domain and may optionally lack all or part of the transmembrane domains. The decoy receptor may optionally be fused to an immunoglobulin constant region, e.g. IgG Fc region.

Inhibitors

The present invention contemplates the use of inhibitor molecules capable of binding to one or more of IL-11 , an IL-11 containing complex, IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130, and inhibiting IL-11 mediated signalling.

In some embodiments the agent is a peptide- or polypeptide-based binding agent based on IL-11 , e.g. mutant, variant or binding fragment of IL-11. Suitable peptide or polypeptide based agents may bind to a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130) in a manner that does not lead to initiation of signal transduction, or which produces sub-optimal signalling. IL-11 mutants of this kind may act as competitive inhibitors of endogenous IL-11.

For example, W147A is an IL-11 antagonist in which the amino acid 147 is mutated from a tryptophan to an alanine, which destroys the so-called ‘site III’ of IL-11. This mutant can bind to IL-11 Ra, but engagement of the gp130 homodimer fails, resulting in efficient blockade of IL-11 signalling (Underhill- Day eta!., 2003; Endocrinology 2003 Aug;144(8):3406-14). Lee et al (Am J respire Cell Mol Biol. 2008 Dec; 39(6):739-746) also report the generation of an IL-11 antagonist mutant (a “mutein”) capable of specifically inhibiting the binding of IL-11 to IL-11Ra. IL-11 muteins are also described in WO 2009/052588 A1.

Menkhorst et al (Biology of Reproduction May 1 , 2009 vol.80 no.5920-927) describe a PEGylated IL-11 antagonist, PEGIL11A (CSL Limited, Parkvill, Victoria, Australia) which is effective to inhibit IL-11 action in female mice.

Pasqualini etal. Cancer (2015) 121 (14):2411-2421 describe a ligand-directed, peptidomimetic drug, bone metastasis-targeting peptidomimetic-11 (BMTP-11) capable of binding to IL-11Ra.

In some embodiments a binding agent capable of binding to a receptor for IL-11 may be provided in the form of a small molecule inhibitor of one of IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130. In some embodiments a binding agent may be provided in the form of a small molecule inhibitor of IL-11 or an IL-11 containing complex, e.g. IL-11 inhibitor described in Lay etal., Int. J. Oncol. (2012); 41(2): 759-764, which is hereby incorporated by reference in its entirety.

Aptamers

In some embodiments, an agent capable of binding to IL-11/an IL-11 containing complex or a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130) is an aptamer. Aptamers, also called nucleic acid/peptide ligands, are nucleic acid or peptide molecules characterised by the ability to bind to a target molecule with high specificity and high affinity. Almost every aptamer identified to date is a non-naturally occurring molecule.

Aptamers to a given target (e.g. IL-11 , an IL-11 containing complex or a receptor for IL-11) may be identified and/or produced by the method of Systematic Evolution of Ligands by Exponential enrichment (SELEXTM), or by developing SOMAmers (slow off-rate modified aptamers) (Gold L etal. (2010) PLoS ONE 5(12):e15004). Aptamers and SELEX are described in Tuerk and Gold, Science (1990) 249(4968):505-10, and in WO 91/19813. Applying the SELEX and the SOMAmer technology includes for instance adding functional groups that mimic amino acid side chains to expand the aptamer's chemical diversity. As a result high affinity aptamers for a target may be enriched and identified.

Aptamers may be DNA or RNA molecules and may be single stranded or double stranded. The aptamer may comprise chemically modified nucleic acids, for example in which the sugar and/or phosphate and/or base is chemically modified. Such modifications may improve the stability of the aptamer or make the aptamer more resistant to degradation and may include modification at the 2' position of ribose.

Aptamers may be synthesised by methods which are well known to the skilled person. For example, aptamers may be chemically synthesised, e.g. on a solid support. Solid phase synthesis may use phosphoramidite chemistry. Briefly, a solid supported nucleotide is detritylated, then coupled with a suitably activated nucleoside phosphoramidite to form a phosphite triester linkage. Capping may then occur, followed by oxidation of the phosphite triester with an oxidant, typically iodine. The cycle may then be repeated to assemble the aptamer (e.g., see Sinha, N. D.; Biernat, J.; McManus, J.; Koster, H. Nucleic Acids Res. 1984, 12, 4539; and Beaucage, S. L; Lyer, R. P. (1992). Tetrahedron 48 (12): 2223).

Suitable nucleic acid aptamers may optionally have a minimum length of one of 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides. Suitable nucleic acid aptamers may optionally have a maximum length of one of 20, 21 , 22, 23, 24, 25,

26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 , 52, 53,

54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, or 80 nucleotides. Suitable nucleic acid aptamers may optionally have a length of one of 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43,

44, 45, 46, 47, 48, 49, 50, 51 , 52, 53, 54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 ,

72, 73, 74, 75, 76, 77, 78, 79, or 80 nucleotides.

Aptamers may be peptides selected or engineered to bind specific target molecules. Peptide aptamers and methods for their generation and identification are reviewed in Reverdatto et at., Curr Top Med Chem. (2015) 15(12):1082-101 , which is hereby incorporated by reference in its entirety. Peptide aptamers may optionally have a minimum length of one of 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acids. Peptide aptamers may optionally have a maximum length of one of 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26,

27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49 or 50 amino acids. Suitable peptide aptamers may optionally have a length of one of 2-30, 2-25, 2-20, 5-30, 5-25 or 5-20 amino acids.

Aptamers may have KD’S in the nM or pM range, e.g. less than one of 500nM, 10OnM, 50nM, 10nM, 1 nM, 500pM, 100pM.

Properties of I L- 11 binding agents

Agents capable of binding to IL-11/an IL-11 containing complex or a receptor for IL-11 according to the present invention may exhibit one or more of the following properties:

• Specific binding to IL-11 /IL-11 containing complex or a receptor for IL-11 ;

• Binding to IL-11 /IL-11 containing complex, or a receptor for IL-11 , with a KD of 10pM or less, preferably one of < 5pM < 1 pM, <500nM, < 10OnM, <1 OnM, <1 nM or <1 OOpM;

• Inhibition of interaction between IL-11 and IL-11Ra;

• Inhibition of interaction between IL-11 and gp130; • Inhibition of interaction between IL-11 and IL-11 Rcrgpl 30 receptor complex;

• Inhibition of interaction between IL-11 : 1 L- 11 Ra complex and gp130; and

• Inhibition of interaction between IL-11:IL-11 Rcrgpl 30 complexes (i.e. multimerisation of such complexes).

These properties can be determined by analysis of the relevant agent in a suitable assay, which may involve comparison of the performance of the agent to suitable control agents. The skilled person is able to identify an appropriate control conditions for a given assay.

For example, a suitable negative control for the analysis of the ability of a test antibody/antigen-binding fragment to bind to IL-11/an IL-11 containing complex/a receptor for IL-11 may be an antibody/antigenbinding fragment directed against a non-target protein (i.e. an antibody/antigen-binding fragment which is not specific for IL-11/an IL-11 containing complex/a receptor for IL-11). A suitable positive control may be a known, validated (e.g. commercially available) IL-11 - or IL-11 receptor-binding antibody. Controls may be of the same isotype as the putative IL-11/IL-11 containing complex/IL-11 receptor-binding antibody/antigen-binding fragment being analysed, and may e.g. have the same constant regions.

In some embodiments, the agent may be capable of binding specifically to IL-11 or an IL-11 containing complex, or a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130). An agent which specifically binds to a given target molecule preferably binds the target with greater affinity, and/or with greater duration than it binds to other, non-target molecules.

In some embodiments the agent may bind to IL-11 or an IL-11 containing complex with greater affinity than the affinity of binding to one or more other members of the IL-6 cytokine family (e.g. IL-6, leukemia inhibitory factor (LIF), oncostatin M (OSM), cardiotrophin-1 (CT-1), ciliary neurotrophic factor (CNTF) and cardiotrophin-like cytokine (CLC)). In some embodiments the agent may bind to a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11Ra and/or gp130) with greater affinity than the affinity of binding to one or more other members of the IL-6 receptor family. In some embodiments the agent may bind with greater affinity to IL-11 Ra than the affinity of binding to one or more of IL-6Ra, leukemia inhibitory factor receptor (LIFR), oncostatin M receptor (OSMR), ciliary neurotrophic factor receptor alpha (CNTFRa) and cytokine receptor-like factor 1 (CRLF1).

In some embodiments, the extent of binding of a binding agent to an non-target is less than about 10% of the binding of the agent to the target as measured, e.g., by ELISA, SPR, Bio-Layer Interferometry (BLI), MicroScale Thermophoresis (MST), or by a radioimmunoassay (RIA). Alternatively, the binding specificity may be reflected in terms of binding affinity, where the binding agent binds to IL-11 , an IL-11 containing complex or a receptor for IL-11 with a KD that is at least 0.1 order of magnitude (i.e. 0.1 x 10n, where n is an integer representing the order of magnitude) greater than the KD towards another, non-target molecule. This may optionally be one of at least 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, or 2.0. Binding affinity for a given binding agent for its target is often described in terms of its dissociation constant (KD). Binding affinity can be measured by methods known in the art, such as by ELISA, Surface Plasmon Resonance (SPR; see e.g. Hearty eta!., Methods Mol Biol (2012) 907:411-442; or Rich et a!., Anal Biochem. 2008 Feb 1; 373(1 ): 112-20), Bio-Layer Interferometry (see e.g. Lad et al., (2015) J Biomol Screen 20(4): 498-507; or Concepcion et al., Comb Chem High Throughput Screen. 2009 Sep; 12(8)791-800), MicroScale Thermophoresis (MST) analysis (see e.g. Jerabek-Willemsen et al., Assay Drug Dev Technol. 2011 Aug; 9(4): 342-353), or by a radiolabelled antigen binding assay (RIA).

In some embodiments, the agent is capable of binding to IL-11 or an IL-11 containing complex, or a receptor for IL-11 with a KD of 50 pM or less, preferably one of <10 pM, <5 pM, <4 pM, <3 pM, <2 pM, <1 pM, <500 nM, <100 nM, <75 nM, <50 nM, <40 nM, <30 nM, <20 nM, <15 nM, <12.5 nM, <10 nM, <9 nM, <8 nM, <7 nM, <6 nM, <5 nM, <4 nM <3 nM, <2 nM, <1 nM, <500 pM, <400 pM, <300 pM, <200 pM, or <100 pM.

In some embodiments, the agent binds to IL-11 , an IL-11 containing complex or a receptor for IL-11 with an affinity of binding (e.g. as determined by ELISA) of EC50 = 10,000 ng/ml or less, preferably one of <5,000 ng/ml, <1000 ng/ml, <900 ng/ml, <800 ng/ml, <700 ng/ml, <600 ng/ml, <500 ng/ml, <400 ng/ml, <300 ng/ml, <200 ng/ml, <100 ng/ml, <90 ng/ml, <80 ng/ml, <70 ng/ml, <60 ng/ml, <50 ng/ml, <40 ng/ml, <30 ng/ml, <20 ng/ml, <15 ng/ml, <10 ng/ml, <7.5 ng/ml, <5 ng/ml, <2.5 ng/ml, or<1 ng/ml. Such ELISAs can be performed e.g. as described in Antibody Engineering, vol. 1 (2nd Edn), Springer Protocols, Springer (2010), Part V, pp657-665.

In some embodiments, the agent binds to IL-11 or an IL-11 -containing complex in a region which is important for binding to a receptor for the IL-11 or IL-11 -containing complex, e.g. gp130 or IL-11 Ra, and thereby inhibits interaction between IL-11 or an IL-11 -containing complex and a receptor for IL-11 , and/or signalling through the receptor. In some embodiments, the agent binds to a receptor for IL-11 in a region which is important for binding to IL-11 or an IL-11 -containing complex, and thereby inhibits interaction between IL-11 or an IL-11 -containing complex and a receptor for IL-11 , and/or signalling through the receptor.

The ability of a given binding agent (e.g. an agent capable of binding IL-11/an IL-11 containing complex or a receptor for IL-11) to inhibit interaction between two proteins can be determined for example by analysis of interaction in the presence of, or following incubation of one or both of the interaction partners with, the binding agent. An example of a suitable assay to determine whether a given binding agent is capable of inhibiting interaction between two interaction partners is a competition ELISA.

A binding agent which is capable of inhibiting a given interaction (e.g. between IL-11 and IL-11 Ra, or between IL-11 and gp130, or between IL-11 and IL-11 Rcrgpl 30, or between IL-11 : 1 L- 11 Ra and gp130, or between IL-11 :IL-11 Ra:gp130 complexes) is identified by the observation of a reduction/decrease in the level of interaction between the interaction partners in the presence of - or following incubation of one or both of the interaction partners with - the binding agent, as compared to the level of interaction in the absence of the binding agent (or in the presence of an appropriate control binding agent). Suitable analysis can be performed in vitro, e.g. using recombinant interaction partners or using cells expressing the interaction partners. Cells expressing interaction partners may do so endogenously, or may do so from nucleic acid introduced into the cell. For the purposes of such assays, one or both of the interaction partners and/or the binding agent may be labelled or used in conjunction with a detectable entity for the purposes of detecting and/or measuring the level of interaction. For example, the agent may be labelled with a radioactive atom or a coloured molecule or a fluorescent molecule or a molecule which can be readily detected in any other way. Suitable detectable molecules include fluorescent proteins, luciferase, enzyme substrates, and radiolabels. The binding agent may be directly labelled with a detectable label or it may be indirectly labelled. For example, the binding agent may be unlabelled, and detected by another binding agent which is itself labelled. Alternatively, the second binding agent may have bound to it biotin and binding of labelled streptavidin to the biotin may be used to indirectly label the first binding agent.

Ability of a binding agent to inhibit interaction between two binding partners can also be determined by analysis of the downstream functional consequences of such interaction, e.g. IL-11 -mediated signalling. For example, downstream functional consequences of interaction between IL-11 and IL-11Ra:gp130 or between IL-11 : 1 L- 11 Ra and gp130, or between IL-11 :IL-11 Rccgpl 30 complexes may include e.g. a process mediated by IL-11 , or gene/protein expression of e.g. collagen or IL-11.

Inhibition of interaction between IL-11 or an IL-11 containing complex and a receptor for IL-11 can be analysed using 3H-thymidine incorporation and/or Ba/F3 cell proliferation assays such as those described in e.g. Curtis etal. Blood, 1997, 90(11) and Karpovich etal. Mol. Hum. Reprod. 2003 9(2): 75-80. Ba/F3 cells co-express IL-11Ra and gp130.

In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 and IL- 11 Ra to less than 100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less,

30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less of the level of interaction between IL-11 and IL-11Ra in the absence of the binding agent (or in the presence of an appropriate control binding agent). In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 and IL-11 Ra to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of interaction between IL-11 and IL-11 Ra in the absence of the binding agent (or in the presence of an appropriate control binding agent).

In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 and gp130 to less than 100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less,

30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less of the level of interaction between IL-11 and gp130 in the absence of the binding agent (or in the presence of an appropriate control binding agent). In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 and gp130 to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of interaction between IL-11 and gp130 in the absence of the binding agent (or in the presence of an appropriate control binding agent).

In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 and IL- 11 Rcrgpl 30 to less than 100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less of the level of interaction between IL-11 and IL-11 Rcrgpl 30 in the absence of the binding agent (or in the presence of an appropriate control binding agent). In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 and IL-11 Rcrgpl 30 to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of interaction between IL-11 and IL-11 Rcrgpl 30 in the absence of the binding agent (or in the presence of an appropriate control binding agent).

In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 :IL-11Ra complex and gp130 to less than 100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less of the level of interaction between IL-11 : IL-11 Ra complex and gp130 in the absence of the binding agent (or in the presence of an appropriate control binding agent). In some embodiments, the binding agent is capable of inhibiting interaction between IL-11 :IL-11 Ra complex and gp130 to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of interaction between IL-11 : 1 L- 11 Ra complex and gp130 in the absence of the binding agent.

In some embodiments, the binding agent may be capable of inhibiting interaction between IL-11 : 1 L- 11 Ra:gp130 complexes (i.e. multimerisation of such complexes) to less than 100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less ofthe level of interaction between IL-11 :IL-11 Ra:gp130 complexes in the absence ofthe binding agent (or in the presence of an appropriate control binding agent). In some embodiments, the binding agent is capable of inhibiting interaction between IL-11 : 1 L- 11 Ra:gp130 complexes to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of interaction between IL-11 :IL-11 Ra:gp130 complexes in the absence of the binding agent.

Agents capable of reducing expression of IL-11 or an IL-11 receptor In aspects of the present invention the agent capable of inhibiting IL-11 -mediated signalling may be capable of preventing or reducing the expression of one or more of IL-11 , IL-11 Ra or gp130.

Expression may be gene or protein expression, and may be determined as described herein or by methods in the art that will be well known to a skilled person. Expression may be by a cell/tissue/organ/organ system of a subject.

Suitable agents may be of any kind, but in some embodiments an agent capable of preventing or reducing the expression of one or more of IL-11 , IL-11 Ra or gp130 may be a small molecule or an oligonucleotide.

An agent capable of preventing or reducing of the expression of one or more of IL-11 , IL-11Ra or gp130 may do so e.g. through inhibiting transcription of the gene encoding IL-11 , IL-11Ra or gp130, inhibiting post-transcriptional processing of RNA encoding IL-11 , IL-11 Ra or gp130, reducing the stability of RNA encoding IL-11 , IL-11 Ra or gp130, promoting degradation of RNA encoding IL-11 , IL-11 Ra or gp130, inhibiting post-translational processing of IL-11 , IL-11 Ra or gp130 polypeptide, reducing the stability of IL- 11 , IL-11 Ra or gp130 polypeptide or promoting degradation of IL-11 , IL-11 Ra or gp130 polypeptide.

Taki etal. Clin Exp Immunol (1998) Apr; 112(1): 133-138 reported a reduction in the expression of IL-11 in rheumatoid synovial cells upon treatment with indomethacin, dexamethasone or interferon-gamma (IFNy).

The present invention contemplates the use of antisense nucleic acid to prevent/reduce expression of IL- 11 , IL-11 Ra or gp130. In some embodiments, an agent capable of preventing or reducing the expression of IL-11 , IL-11 Ra or gp130 may cause reduced expression by RNA interference (RNAi).

In some embodiments, the agent may be an inhibitory nucleic acid, such as antisense or small interfering RNA, including but not limited to shRNA orsiRNA.

In some embodiments the inhibitory nucleic acid is provided in a vector. For example, in some embodiments the agent may be a lentiviral vector encoding shRNA for one or more of IL-11 , IL-11 Ra or gp130.

Oligonucleotide molecules, particularly RNA, may be employed to regulate gene expression. These include antisense oligonucleotides, targeted degradation of mRNAs by small interfering RNAs (siRNAs), post transcriptional gene silencing (PTGs), developmental^ regulated sequence-specific translational repression of mRNA by micro-RNAs (miRNAs) and targeted transcriptional gene silencing. An antisense oligonucleotide is an oligonucleotide, preferably single-stranded, that targets and binds, by complementary sequence binding, to a target oligonucleotide, e.g. mRNA. Where the target oligonucleotide is an mRNA, binding of the antisense to the mRNA blocks translation of the mRNA and expression of the gene product. Antisense oligonucleotides may be designed to bind sense genomic nucleic acid and inhibit transcription of a target nucleotide sequence.

In view of the known nucleic acid sequences for IL-11 , IL-11Ra and gp130 (e.g. the known mRNA sequences available from GenBank under Accession No.s: BC012506.1 Gl:15341754 (human IL-11 ) ,

BC134354.1 Gl:126632002 (mouse IL-11), AF347935.1 GL13549072 (rat IL-11), NM_001142784.2 Gl:391353394 (human IL-11 Ra), NM_001163401.1 GL254281268 (mouse IL-11 Ra), NIVM39116.1 GL20806172 (rat IL-11 Ra), NM_001190981.1 GL300244534 (human gp130), NM_010560.3 GL225007624 (mouse gp130), NM_001008725.3 GL300244570 (ratgp130)) oligonucleotides may be designed to repress or silence the expression of IL-11 , IL-11 Ra or gp130.

Such oligonucleotides may have any length, but may preferably be short, e.g. less than 100 nucleotides, e.g. 10-40 nucleotides, or 20-50 nucleotides, and may comprise a nucleotide sequence having complete- or near-complementarity (e.g. 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% complementarity) to a sequence of nucleotides of corresponding length in the target oligonucleotide, e.g. the IL-11 , IL-11Ra orgp130 mRNA. The complementary region of the nucleotide sequence may have any length, but is preferably at least 5, and optionally no more than 50, nucleotides long, e.g. one of 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides.

Repression of expression of IL-11 , IL-11 Ra or gp130 will preferably result in a decrease in the quantity of IL-11 , IL-11 Ra or gp130 expressed by a cell/tissue/organ/organ system/subject. For example, in a given cell the repression of IL-11 , IL-11 Ra or gp130 by administration of a suitable nucleic acid will result in a decrease in the quantity of IL-11 , IL-11 Ra or gp130 expressed by that cell relative to an untreated cell. Repression may be partial. Preferred degrees of repression are at least 50%, more preferably one of at least 60%, 70%, 80%, 85% or 90%. A level of repression between 90% and 100% is considered a ‘silencing’ of expression or function.

A role for the RNAi machinery and small RNAs in targeting of heterochromatin complexes and epigenetic gene silencing at specific chromosomal loci has been demonstrated. Double-stranded RNA (dsRNA)- dependent post transcriptional silencing, also known as RNA interference (RNAi), is a phenomenon in which dsRNA complexes can target specific genes of homology for silencing in a short period of time. It acts as a signal to promote degradation of mRNA with sequence identity. A 20-nt siRNA is generally long enough to induce gene-specific silencing, but short enough to evade host response. The decrease in expression of targeted gene products can be extensive with 90% silencing induced by a few molecules of siRNA. RNAi based therapeutics have been progressed into Phase I, II and III clinical trials fora number of indications (Nature 2009 Jan 22; 457(7228) :426-433). In the art, these RNA sequences are termed "short or small interfering RNAs" (siRNAs) or "microRNAs" (miRNAs) depending on their origin. Both types of sequence may be used to down-regulate gene expression by binding to complementary RNAs and either triggering mRNA elimination (RNAi) or arresting mRNA translation into protein. siRNA are derived by processing of long double stranded RNAs and when found in nature are typically of exogenous origin. Micro-interfering RNAs (miRNA) are endogenously encoded small non-coding RNAs, derived by processing of short hairpins. Both siRNA and miRNA can inhibit the translation of mRNAs bearing partially complimentary target sequences without RNA cleavage and degrade mRNAs bearing fully complementary sequences. siRNA ligands are typically double stranded and, in order to optimise the effectiveness of RNA mediated down-regulation of the function of a target gene, it is preferred that the length of the siRNA molecule is chosen to ensure correct recognition of the siRNA by the RISC complex that mediates the recognition by the siRNA of the mRNA target and so that the siRNA is short enough to reduce a host response. miRNA ligands are typically single stranded and have regions that are partially complementary enabling the ligands to form a hairpin. miRNAs are RNA genes which are transcribed from DNA, but are not translated into protein. A DNA sequence that codes for a miRNA gene is longer than the miRNA. This DNA sequence includes the miRNA sequence and an approximate reverse complement. When this DNA sequence is transcribed into a single-stranded RNA molecule, the miRNA sequence and its reverse- complement base pair to form a partially double stranded RNA segment. The design of microRNA sequences is discussed in John et al, PLoS Biology, 11(2), 1862-1879, 2004.

Typically, the RNA ligands intended to mimic the effects of siRNA or miRNA have between 10 and 40 ribonucleotides (or synthetic analogues thereof), more preferably between 17 and 30 ribonucleotides, more preferably between 19 and 25 ribonucleotides and most preferably between 21 and 23 ribonucleotides. In some embodiments of the invention employing double-stranded siRNA, the molecule may have symmetric 3' overhangs, e.g. of one or two (ribo)nucleotides, typically a UU of dTdT 3' overhang. Based on the disclosure provided herein, the skilled person can readily design suitable siRNA and miRNA sequences, for example using resources such the Ambion siRNA finder. siRNA and miRNA sequences can be synthetically produced and added exogenously to cause gene downregulation or produced using expression systems (e.g. vectors). In a preferred embodiment the siRNA is synthesized synthetically.

Longer double stranded RNAs may be processed in the cell to produce siRNAs (see for example Myers (2003) Nature Biotechnology 21 :324-328). The longer dsRNA molecule may have symmetric 3' or 5' overhangs, e.g. of one or two (ribo)nucleotides, or may have blunt ends. The longer dsRNA molecules may be 25 nucleotides or longer. Preferably, the longer dsRNA molecules are between 25 and 30 nucleotides long. More preferably, the longer dsRNA molecules are between 25 and 27 nucleotides long. Most preferably, the longer dsRNA molecules are 27 nucleotides in length. dsRNAs 30 nucleotides or more in length may be expressed using the vector pDECAP (Shinagawa etai, Genes and Dev., 17, 1340-5, 2003).

Another alternative is the expression of a short hairpin RNA molecule (shRNA) in the cell. shRNAs are more stable than synthetic siRNAs. A shRNA consists of short inverted repeats separated by a small loop sequence. One inverted repeat is complimentary to the gene target. In the cell the shRNA is processed by DICER into a siRNA which degrades the target gene mRNA and suppresses expression. In a preferred embodiment the shRNA is produced endogenously (within a cell) by transcription from a vector. shRNAs may be produced within a cell by transfecting the cell with a vector encoding the shRNA sequence under control of a RNA polymerase III promoter such as the human H1 or 7SK promoter or a RNA polymerase II promoter. Alternatively, the shRNA may be synthesised exogenously (in vitro) by transcription from a vector. The shRNA may then be introduced directly into the cell. Preferably, the shRNA molecule comprises a partial sequence of IL-11 , IL-11 Ra or gp130. Preferably, the shRNA sequence is between 40 and 100 bases in length, more preferably between 40 and 70 bases in length. The stem of the hairpin is preferably between 19 and 30 base pairs in length. The stem may contain G-U pairings to stabilise the hairpin structure. siRNA molecules, longer dsRNA molecules or miRNA molecules may be made recombinantly by transcription of a nucleic acid sequence, preferably contained within a vector. Preferably, the siRNA molecule, longer dsRNA molecule or miRNA molecule comprises a partial sequence of IL-11 , IL-11 Ra or gp130.

In one embodiment, the siRNA, longer dsRNA or miRNA is produced endogenously (within a cell) by transcription from a vector. The vector may be introduced into the cell in any of the ways known in the art. Optionally, expression of the RNA sequence can be regulated using a tissue specific (e.g. liver specific) promoter. In a further embodiment, the siRNA, longer dsRNA or miRNA is produced exogenously (in vitro) by transcription from a vector.

Suitable vectors may be oligonucleotide vectors configured to express the oligonucleotide agent capable of IL-11 , IL-11 Ra or gp130 repression. Such vectors may be viral vectors or plasmid vectors. The therapeutic oligonucleotide may be incorporated in the genome of a viral vector and be operably linked to a regulatory sequence, e.g. promoter, which drives its expression. The term “operably linked” may include the situation where a selected nucleotide sequence and regulatory nucleotide sequence are covalently linked in such a way as to place the expression of a nucleotide sequence under the influence or control of the regulatory sequence. Thus a regulatory sequence is operably linked to a selected nucleotide sequence if the regulatory sequence is capable of effecting transcription of a nucleotide sequence which forms part or all of the selected nucleotide sequence.

Viral vectors encoding promoter-expressed siRNA sequences are known in the art and have the benefit of long term expression of the therapeutic oligonucleotide. Examples include lentiviral ( Nature 2009 Jan 22; 457(7228) :426-433), adenovirus (Shen etal., FEBS Lett 2003 Mar 27;539(1 -3)111-4) and retroviruses (Barton and Medzhitov PNAS November 12, 2002 vol.99, no.23 14943-14945).

In other embodiments a vector may be configured to assist delivery of the therapeutic oligonucleotide to the site at which repression of IL-11 , IL-11Ra or gp130 expression is required. Such vectors typically involve complexing the oligonucleotide with a positively charged vector (e.g., cationic cell penetrating peptides, cationic polymers and dendrimers, and cationic lipids); conjugating the oligonucleotide with small molecules (e.g., cholesterol, bile acids, and lipids), polymers, antibodies, and RNAs; or encapsulating the oligonucleotide in nanoparticulate formulations (Wang etai, AAPS J. 2010 Dec; 12(4): 492-503).

In one embodiment, a vector may comprise a nucleic acid sequence in both the sense and antisense orientation, such that when expressed as RNA the sense and antisense sections will associate to form a double stranded RNA.

Alternatively, siRNA molecules may be synthesized using standard solid or solution phase synthesis techniques which are known in the art. Linkages between nucleotides may be phosphodiester bonds or alternatives, for example, linking groups of the formula P(0)S, (thioate); P(S)S, (dithioate); P(0)NR'2; P(0)R'; P(0)0R6; CO; or CONR'2 wherein R is H (or a salt) or alkyl (1-12C) and R6 is alkyl (1-9C) is joined to adjacent nucleotides through-O-or-S-.

Modified nucleotide bases can be used in addition to the naturally occurring bases, and may confer advantageous properties on siRNA molecules containing them.

For example, modified bases may increase the stability of the siRNA molecule, thereby reducing the amount required for silencing. The provision of modified bases may also provide siRNA molecules which are more, or less, stable than unmodified siRNA.

The term ‘modified nucleotide base’ encompasses nucleotides with a covalently modified base and/or sugar. For example, modified nucleotides include nucleotides having sugars which are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3'position and other than a phosphate group at the 5'position. Thus modified nucleotides may also include 2'substituted sugars such as 2'-0-methyl- ; 2'-0-alkyl ; 2'-0-allyl ; 2'-S-alkyl; 2'-S-allyl; 2'-fluoro- ; 2'-halo or azido- ribose, carbocyclic sugar analogues, a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, and sedoheptulose.

Modified nucleotides are known in the art and include alkylated purines and pyrimidines, acylated purines and pyrimidines, and other heterocycles. These classes of pyrimidines and purines are known in the art and include pseudoisocytosine, N4,N4-ethanocytosine, 8-hydroxy-N6-methyladenine, 4-acetylcytosine,5- (carboxyhydroxylmethyl) uracil, 5 fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5- carboxymethylaminomethyl uracil, dihydrouracil, inosine, N6-isopentyl-adenine, 1-methyladenine, 1- methylpseudouracil, 1-methylguanine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3- methylcytosine, 5-methylcytosine, N6-methyladenine, 7-methylguanine, 5-methylaminomethyl uracil, 5- methoxy amino methyl-2-thiouracil, -D-mannosylqueosine, 5-methoxycarbonylmethyluracil, 5methoxyuracil, 2 methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid methyl ester, pseudouracil, 2-thiocytosine, 5-methyl-2 thiouracil, 2-thiouracil, 4-thiouracil, 5methyluracil, N-uracil-5-oxyacetic acid methylester, uracil 5-oxyacetic acid, queosine, 2-thiocytosine, 5-propyluracil, 5-propylcytosine, 5- ethyluracil, 5ethylcytosine, 5-butyluracil, 5-pentyluracil, 5-pentylcytosine, and 2,6,diaminopurine, methylpsuedouracil, 1-methylguanine, 1 -methylcytosine.

Methods relating to the use of RNAi to silence genes in C. elegans, Drosophila, plants, and mammals are known in the art (Fire A, et al., 1998 Nature 391:806-811; Fire, A. Trends Genet. 15, 358-363 (1999); Sharp, P. A. RNA interference 2001. Genes Dev. 15, 485-490 (2001); Hammond, S. M., et al., Nature Rev. Genet. 2, 110-1119 (2001); Tuschl, T. Chem. Biochem. 2, 239-245 (2001); Hamilton, A. etal., Science 286, 950-952 (1999); Hammond, S. M., etal., Nature 404, 293-296 (2000); Zamore, P. D., etal., Cell 101, 25-33 (2000); Bernstein, E., etal., Nature 409, 363-366 (2001); Elbashir, S. M., etal., Genes Dev. 15, 188-200 (2001); WO0129058; W09932619, and Elbashir S M, etal., 2001 Nature 411 :494-498).

Accordingly, the invention provides nucleic acid that is capable, when suitably introduced into or expressed within a mammalian, e.g. human, cell that otherwise expresses IL-11 , IL-11 Ra or gp130, of suppressing IL-11 , IL-11 Ra or gp130 expression by RNAi.

Nucleic acid sequences for IL-11 , IL-11 Ra and gp130 (e.g. the known mRNA sequences available from GenBank under Accession No.s: BC012506.1 GL15341754 (human IL-11), BC134354.1 GL126632002 (mouse IL-11), AF347935.1 GL13549072 (rat IL-11), NM_001142784.2 GL391353394 (human IL-11 Ra), NM_001163401.1 GL254281268 (mouse IL-11 Ra), NM 39116.1 GL20806172 (rat IL-11 Ra),

NM_001190981.1 GL300244534 (human gp130), NM_010560.3 GL225007624 (mouse gp130),

NM_001008725.3 GL300244570 (rat gp130)) oligonucleotides may be designed to repress or silence the expression of IL-11 , IL-11 Ra or gp130.

The nucleic acid may have substantial sequence identity to a portion of IL-11 , IL-11 Ra or gp130 mRNA, e.g. as defined in GenBank accession no. NM_000641.3 Gl:391353405 (IL-11), NM_001142784.2 GL391353394 (IL-11 Ra), NM_001190981.1 Gl:300244534 (gp130) or the complementary sequence to said mRNA.

The nucleic acid may be a double-stranded siRNA. (As the skilled person will appreciate, and as explained further below, a siRNA molecule may include a short 3’ DNA sequence also.)

Alternatively, the nucleic acid may be a DNA (usually double-stranded DNA) which, when transcribed in a mammalian cell, yields an RNA having two complementary portions joined via a spacer, such that the RNA takes the form of a hairpin when the complementary portions hybridise with each other. In a mammalian cell, the hairpin structure may be cleaved from the molecule by the enzyme DICER, to yield two distinct, but hybridised, RNA molecules.

In some preferred embodiments, the nucleic acid is generally targeted to the sequence of one of SEQ ID NOs 4 to 7 (IL-11) or to one of SEQ ID NOs 8 to 11 (IL-11 Ra).

Only single-stranded (i.e. non self-hybridised) regions of an mRNA transcript are expected to be suitable targets for RNAi. It is therefore proposed that other sequences very close in the IL-11 or IL-11 Ra mRNA transcript to the sequence represented by one of SEQ ID NOs 4 to 7 or 8 to 11 may also be suitable targets for RNAi. Such target sequences are preferably 17-23 nucleotides in length and preferably overlap one of SEQ ID NOs 4 to 7 or 8 to 11 by at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18 or all 19 nucleotides (at either end of one of SEQ ID NOs 4 to 7 or 8 to 11).

Accordingly, the invention provides nucleic acid that is capable, when suitably introduced into or expressed within a mammalian cell that otherwise expresses IL-11 or IL-11 Ra, of suppressing IL-11 or IL- 11 Ra expression by RNAi, wherein the nucleic acid is generally targeted to the sequence of one of SEQ ID NOs 4 to 7 or 8 to 11.

By “generally targeted” the nucleic acid may target a sequence that overlaps with SEQ ID NOs 4 to 7 or 8 to 11 . In particular, the nucleic acid may target a sequence in the mRNA of human IL-11 or IL-11 Ra that is slightly longer or shorter than one of SEQ ID NOs 4 to 7 or 8 to 11 (preferably from 17-23 nucleotides in length), but is otherwise identical to one of SEQ ID NOs 4 to 7 or 8 to 11.

It is expected that perfect identity/complementarity between the nucleic acid of the invention and the target sequence, although preferred, is not essential. Accordingly, the nucleic acid of the invention may include a single mismatch compared to the mRNA of IL-11 or IL-11 Ra. It is expected, however, that the presence of even a single mismatch is likely to lead to reduced efficiency, so the absence of mismatches is preferred. When present, 3’ overhangs may be excluded from the consideration of the number of mismatches.

The term “complementarity” is not limited to conventional base pairing between nucleic acid consisting of naturally occurring ribo- and/or deoxyribonucleotides, but also includes base pairing between mRNA and nucleic acids of the invention that include non-natural nucleotides.

In one embodiment, the nucleic acid (herein referred to as double-stranded siRNA) includes the double- stranded RNA sequences shown in SEQ ID NOs 12 to 15. In another embodiment, the nucleic acid (herein referred to as double-stranded siRNA) includes the double-stranded RNA sequences shown in SEQ ID NOs 16 to 19. However, it is also expected that slightly shorter or longer sequences directed to the same region of IL-11 or IL-11 Ra mRNA will also be effective. In particular, it is expected that double-stranded sequences between 17 and 23 bp in length will also be effective.

The strands that form the double-stranded RNA may have short 3’ dinucleotide overhangs, which may be DNA or RNA. The use of a 3’ DNA overhang has no effect on siRNA activity compared to a 3’ RNA overhang, but reduces the cost of chemical synthesis of the nucleic acid strands (Elbashir etai, 2001c). For this reason, DNA dinucleotides may be preferred.

When present, the dinucleotide overhangs may be symmetrical to each other, though this is not essential. Indeed, the 3’ overhang of the sense (upper) strand is irrelevant for RNAi activity, as it does not participate in mRNA recognition and degradation (Elbashir et at., 2001a, 2001b, 2001c).

While RNAi experiments in Drosophila show that antisense 3’ overhangs may participate in mRNA recognition and targeting (Elbashir etai 2001c), 3’ overhangs do not appear to be necessary for RNAi activity of siRNA in mammalian cells. Incorrect annealing of 3’ overhangs is therefore thought to have little effect in mammalian cells (Elbashir etai. 2001c; Czauderna etai. 2003).

Any dinucleotide overhang may therefore be used in the antisense strand of the siRNA. Nevertheless, the dinucleotide is preferably -UU or-UG (or-TT or-TG if the overhang is DNA), more preferably -UU (or- TT). The -UU (or-TT) dinucleotide overhang is most effective and is consistent with (i.e. capable of forming part of) the RNA polymerase III end of transcription signal (the terminator signal is TTTTT). Accordingly, this dinucleotide is most preferred. The dinucleotides AA, CC and GG may also be used, but are less effective and consequently less preferred.

Moreover, the 3’ overhangs may be omitted entirely from the siRNA.

The invention also provides single-stranded nucleic acids (herein referred to as single-stranded siRNAs) respectively consisting of a component strand of one of the aforementioned double-stranded nucleic acids, preferably with the 3’-overhangs, but optionally without. The invention also provides kits containing pairs of such single-stranded nucleic acids, which are capable of hybridising with each other in vitro to form the aforementioned double-stranded siRNAs, which may then be introduced into cells.

The invention also provides DNA that, when transcribed in a mammalian cell, yields an RNA (herein also referred to as an shRNA) having two complementary portions which are capable of self-hybridising to produce a double-stranded motif, e.g. including a sequence selected from the group consisting of SEQ ID NOs: 12 to 15 or 16 to 19 or a sequence that differs from any one of the aforementioned sequences by a single base pair substitution.

The complementary portions will generally be joined by a spacer, which has suitable length and sequence to allow the two complementary portions to hybridise with each other. The two complementary (i.e. sense and antisense) portions may be joined 5’-3’ in either order. The spacer will typically be a short sequence, of approximately 4-12 nucleotides, preferably 4-9 nucleotides, more preferably 6-9 nucleotides.

Preferably the 5’ end of the spacer (immediately 3’ of the upstream complementary portion) consists of the nucleotides -UU- or-UG-, again preferably -UU- (though, again, the use of these particular dinucleotides is not essential). A suitable spacer, recommended for use in the pSuper system of OligoEngine (Seattle, Washington, USA) is UUCAAGAGA. In this and other cases, the ends of the spacer may hybridise with each other, e.g. elongating the double-stranded motif beyond the exact sequences of SEQ ID NOs 12 to 15 or 16 to 19 by a small number (e.g. 1 or 2) of base pairs.

Similarly, the transcribed RNA preferably includes a 3’ overhang from the downstream complementary portion. Again, this is preferably -UU or-UG, more preferably -UU.

Such shRNA molecules may then be cleaved in the mammalian cell by the enzyme DICER to yield a double-stranded siRNA as described above, in which one or each strand of the hybridised dsRNA includes a 3’ overhang.

Techniques for the synthesis of the nucleic acids of the invention are of course well known in the art.

The skilled person is well able to construct suitable transcription vectors for the DNA of the invention using well-known techniques and commercially available materials. In particular, the DNA will be associated with control sequences, including a promoter and a transcription termination sequence.

Of particular suitability are the commercially available pSuper and pSuperior systems of OligoEngine (Seattle, Washington, USA). These use a polymerase-ill promoter (H1) and a T5 transcription terminator sequence that contributes two U residues at the 3’ end of the transcript (which, after DICER processing, provide a 3’ UU overhang of one strand of the siRNA).

Another suitable system is described in Shin etal. (RNA, 2009 May; 15(5): 898-910), which uses another polymerase-ill promoter (U6).

The double-stranded siRNAs of the invention may be introduced into mammalian cells in vitro or in vivo using known techniques, as described below, to suppress expression of IL-11 or a receptor for IL-11.

Similarly, transcription vectors containing the DNAs of the invention may be introduced into tumour cells in vitro or in vivo using known techniques, as described below, for transient or stable expression of RNA, again to suppress expression of IL-11 or a receptor for IL-11.

Accordingly, the invention also provides a method of suppressing expression of IL-11 ora receptor for IL- 11 in a mammalian, e.g. human, cell, the method comprising administering to the cell a double-stranded siRNA of the invention or a transcription vector of the invention. Similarly, the invention further provides a method of treating alcoholic liver disease, comprising administering to a subject a double-stranded siRNA of the invention or a transcription vector of the invention.

The invention further provides the double-stranded siRNAs of the invention and the transcription vectors of the invention, for use in a method of treatment, preferably a method of treating alcoholic liver disease.

The invention further provides the use of the double-stranded siRNAs of the invention and the transcription vectors of the invention in the preparation of a medicament for the treatment of alcoholic liver disease.

The invention further provides a composition comprising a double-stranded siRNA of the invention or a transcription vector of the invention in admixture with one or more pharmaceutically acceptable carriers. Suitable carriers include lipophilic carriers or vesicles, which may assist in penetration of the cell membrane.

Materials and methods suitable for the administration of siRNA duplexes and DNA vectors of the invention are well known in the art and improved methods are under development, given the potential of RNAi technology.

Generally, many techniques are available for introducing nucleic acids into mammalian cells. The choice of technique will depend on whether the nucleic acid is transferred into cultured cells in vitro or in vivo in the cells of a patient. Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE, dextran and calcium phosphate precipitation. In vivo gene transfer techniques include transfection with viral (typically retroviral) vectors and viral coat protein-liposome mediated transfection (Dzau etal. (2003) Trends in Biotechnology 11 , 205-210).

In particular, suitable techniques for cellular administration of the nucleic acids of the invention both in vitro and in vivo are disclosed in the following articles:

General reviews: Borkhardt, A. 2002. Blocking oncogenes in malignant cells by RNA interfere nee-- new hope for a highly specific cancer treatment? Cancer Cell. 2:167-8. Hannon, G.J. 2002. RNA interference. Nature. 418:244-51. McManus, M.T., and P.A. Sharp. 2002. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet. 3:737-47. Scherr, M., M.A. Morgan, and M. Eder. 2003b. Gene silencing mediated by small interfering RNAs in mammalian cells. Curr Med Chem. 10:245-56. Shuey, D.J., D.E. McCallus, and T. Giordano. 2002. RNAi: gene-silencing in therapeutic intervention. Drug Discov Today. 7:1040-6. Systemic delivery using liposomes: Lewis, D.L., J.E. Hagstrom, A.G. Loomis, J.A. Wolff, and H.

Herweijer. 2002. Efficient delivery of siRNA for inhibition of gene expression in postnatal mice. Nat Genet. 32:107-8. Paul, C.P., P.D. Good, I. Winer, and D.R. Engelke. 2002. Effective expression of small interfering RNA in human cells. Nat Biotechnol. 20:505-8. Song, E., S.K. Lee, J. Wang, N. Ince, N. Ouyang, J. Min, J. Chen, P. Shankar, and J. Lieberman. 2003. RNA interference targeting Fas protects mice from fulminant hepatitis. Nat Med. 9:347-51. Sorensen, D.R., M. Leirdal, and M. Sioud. 2003. Gene silencing by systemic delivery of synthetic siRNAs in adult mice. J Mol Biol. 327 :761 -6.

Virus mediated transfer: Abbas-Terki, T., W. Blanco-Bose, N. Deglon, W. Pralong, and P. Aebischer.

2002. Lentiviral-mediated RNA interference. Hum Gene Ther. 13:2197-201. Barton, G.M., and R. Medzhitov. 2002. Retroviral delivery of small interfering RNA into primary cells. Proc Natl Acad Sci U SA. 99:14943-5. Devroe, E., and P.A. Silver. 2002. Retrovirus-delivered siRNA. BMC Biotechnol. 2:15. Lori,

F., P. Guallini, L. Galluzzi, and J. Lisziewicz. 2002. Gene therapy approaches to HIV infection. Am J Pharmacogenomics. 2:245-52. Matta, H., B. Hozayev, R. Tomar, P. Chugh, and P.M. Chaudhary. 2003. Use of lentiviral vectors for delivery of small interfering RNA. Cancer Biol Ther. 2:206-10. Qin, X.F., D.S. An, I.S. Chen, and D. Baltimore. 2003. Inhibiting HIV-1 infection in human T cells by lentiviral-mediated delivery of small interfering RNA against CCR5. Proc Natl Acad Sci U S A. 100:183-8. Scherr, M., K. Battmer, A. Ganser, and M. Eder. 2003a. Modulation of gene expression by lentiviral-mediated delivery of small interfering RNA. Cell Cycle. 2:251-7. Shen, C., A.K. Buck, X. Liu, M. Winkler, and S.N. Reske.

2003. Gene silencing by adenovirus-delivered siRNA. FEBS Lett. 539:111-4.

Peptide delivery: Morris, M.C., L. Chaloin, F. Heitz, and G. Divita. 2000. Translocating peptides and proteins and their use for gene delivery. Curr Opin Biotechnol. 11 :461-6. Simeoni, F., M.C. Morris, F. Heitz, and G. Divita. 2003. Insight into the mechanism of the peptide-based gene delivery system MPG: implications for delivery of siRNA into mammalian cells. Nucleic Acids Res. 31:2717-24. Other technologies that may be suitable for delivery of siRNA to the target cells are based on nanoparticles or nanocapsules such as those described in US patent numbers 6,649,192B and 5,843,509B.

Inhibition of IL-11 -mediated signalling In embodiments of the present invention, agents capable of inhibiting the action of IL-11 may possess one or more of the following functional properties:

• Inhibition of signalling mediated by IL-11 ;

• Inhibition of signalling mediated by binding of IL-11 to IL-11 Rcrgpl 30 receptor complex;

• Inhibition of signalling mediated by binding of IL-11 : 1 L- 11 Ra complex to gp130 (/.e. IL-11 trans signalling);

• Inhibition of signalling mediated by multimerisation of IL-11 : 1 L- 11 Rcrgpl 30 complexes;

• Inhibition of a process mediated by IL-11 ;

• Inhibition of gene/protein expression of IL-11 and/or IL-11 Ra. These properties can be determined by analysis of the relevant agent in a suitable assay, which may involve comparison of the performance of the agent to suitable control agents. The skilled person is able to identify an appropriate control conditions for a given assay.

IL-11 -mediated signalling and/or processes mediated by IL-11 includes signalling mediated by fragments of lL-11 and polypeptide complexes comprising IL-11 or fragments thereof. IL-11 -mediated signalling may be signalling mediated by human IL-11 and/or mouse IL-11. Signalling mediated by IL-11 may occur following binding of IL-11 or an IL-11 containing complex to a receptor to which IL-11 or said complex binds.

In some embodiments, an agent may be capable of inhibiting the biological activity of IL-11 or an IL-11 - containing complex.

In some embodiments, the agent is an antagonist of one or more signalling pathways which are activated by signal transduction through receptors comprising IL-11Ra and/or gp130, e.g. IL-11Rcrgp130. In some embodiments, the agent is capable of inhibiting signalling through one or more immune receptor complexes comprising IL-11Ra and/or gp130, e.g. IL-11Rcrgp130. In various aspects of the present invention, an agent provided herein is capable of inhibiting IL-11 -mediated cis and/or trans signalling. In some embodiments in accordance with the various aspects of the present invention an agent provided herein is capable of inhibiting IL-11 -mediated cis signalling.

In some embodiments, the agent may be capable of inhibiting IL-11 -mediated signalling to less than

100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 80% or less, 75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less of the level of signalling in the absence of the agent (or in the presence of an appropriate control agent). In some embodiments, the agent is capable of reducing IL-11 -mediated signalling to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of signalling in the absence of the agent (or in the presence of an appropriate control agent).

In some embodiments, the IL-11 -mediated signalling may be signalling mediated by binding of IL-11 to IL- 11Rcrgp130 receptor. Such signalling can be analysed e.g. by treating cells expressing IL-11Ra and gp130 with IL-11 , or by stimulating IL-11 production in cells which express IL-11 Ra and gp130.

The IC50 for an agent for inhibition of IL-11 -mediated signalling may be determined, e.g. by culturing Ba/F3 cells expressing IL-11Ra and gp130 in the presence of human IL-11 and the agent, and measuring 3H-thymidine incorporation into DNA. In some embodiments, the agent may exhibit an IC50 of 10 pg/ml or less, preferably one of < 5 pg/ml, < 4 pg/ml, < 3.5 pg/ml, < 3 pg/ml, < 2 pg/ml, < 1 pg/ml, < 0.9 pg/ml, < 0.8 pg/ml, < 0.7 pg/ml, < 0.6 pg/ml, or< 0.5 pg/ml in such an assay. In some embodiments, the IL-11 -mediated signalling may be signalling mediated by binding of IL-11 :IL- 11 Ra complex to gp130. In some embodiments, the IL-11 :IL-11 Ra complex may be soluble, e.g. complex of extracellular domain of IL-11 Ra and IL-11 , or complex of soluble IL-11 Ra isoform/fragment and IL-11.

In some embodiments, the soluble IL-11 Ra is a soluble (secreted) isoform of IL-11 Ra, or is the liberated product of proteolytic cleavage of the extracellular domain of cell membrane bound IL-11Ra.

In some embodiments, the IL-11 : IL-11 Ra complex may be cell-bound, e.g. complex of cell-membrane bound IL-11 Ra and IL-11. Signalling mediated by binding of IL-11 :IL-11 Ra complex to gp130 can be analysed by treating cells expressing gp130 with IL-11 : 1 L- 11 Ra complex, e.g. recombinant fusion protein comprising IL-11 joined by a peptide linker to the extracellular domain of IL-11 Ra, e.g. hyper IL-11. Hyper IL-11 was constructed using fragments of IL-11Ra (amino acid residues 1 to 317 consisting of domain 1 to 3; UniProtKB: Q14626) and IL-11 (amino acid residues 22 to 199 of UniProtKB: P20809) with a 20 amino acid long linker (SEQ ID NO:20). The amino acid sequence for Hyper IL-11 is shown in SEQ ID NO:21.

In some embodiments, the agent may be capable of inhibiting signalling mediated by binding of IL-11 :IL- 11Ra complex to gp130, and is also capable of inhibiting signalling mediated by binding of IL-11 to IL- 11Ra:gp130 receptor.

In some embodiments, the agent may be capable of inhibiting a process mediated by IL-11.

In some embodiments, the agent may be capable of inhibiting gene/protein expression of IL-11 and/or IL- 11 Ra. Gene and/or protein expression can be measured as described herein or by methods in the art that will be well known to a skilled person.

In some embodiments, the agent may be capable of inhibiting gene/protein expression of IL-11 and/or IL- 11 Ra to less than 100%, e.g. one of 99% or less, 95% or less, 90% or less, 85% or less, 80% or less,

75% or less, 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 5% or less, or 1% or less of the level of expression in the absence of the agent (or in the presence of an appropriate control agent). In some embodiments, the agent is capable of inhibiting gene/protein expression of IL-11 and/or IL-11 Ra to less than 1 times, e.g. one of <0.99 times, <0.95 times, <0.9 times, <0.85 times, <0.8 times, <0.75 times, <0.7 times, <0.65 times, <0.6 times, <0.55 times, <0.5 times, <0.45 times, <0.4 times, <0.35 times, <0.3 times, <0.25 times, <0.2 times, <0.15 times, <0.1 times the level of expression in the absence of the agent (or in the presence of an appropriate control agent).

Treatment/prevention of alcoholic liver disease The present invention provides methods and articles (agents and compositions) for the treatment and/or prevention of alcoholic liver disease. Treatment is achieved by inhibition of IL-11 -mediating signalling (i.e. antagonism of IL-11 -mediated signalling). That is, the present invention provides for the treatment/prevention of alcoholic liver disease through inhibition of IL-11 mediated signalling, in e.g. a cell, tissue/organ/organ system/subject. In some embodiments, inhibition of IL-11 -mediated signalling in accordance with the present disclosure comprises inhibition of IL-11 -mediated signalling in the liver, liver tissue and/or cells thereof. In some embodiments, inhibition of IL-11 -mediated signalling in accordance with the present disclosure comprises inhibition of IL- 11 -mediated signalling in hepatic stellate cells and/or hepatocytes.

Accordingly, the present invention provides an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling for use in a method of treating or preventing alcoholic liver disease.

Also provided is the use of an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling for use in the manufacture of a medicament for use in a method of treating or preventing alcoholic liver disease.

Further provided is a method of treating or preventing alcoholic liver disease, the method comprising administering to a subject in need of treatment a therapeutically effective amount of an agent capable of inhibiting interleukin 11 (IL-11)-mediated signalling.

The present invention also provides for the treatment/prevention of diseases/conditions that are caused or exacerbated by alcoholic liver disease. In some embodiments, the present invention provides for the treatment/prevention of diseases/conditions in a subject for which alcoholic liver disease provides a poor prognosis.

In some embodiments, the alcoholic liver disease may be characterised by an increase in the expression of IL-11 and/or IL-11 Ra (i.e. gene and/or protein expression) in the liver/hepatic tissue/liver cells e.g. as compared to the normal level of expression in the relevant organ/tissue/cells (i.e. in the absence of the disease/condition).

Alcoholic liver disease according to the present disclosure may be associated with an upregulation of IL- 11 gene and/or protein expression, e.g. in hepatic cells (e.g. hepatic stellate cells and/or hepatocytes) or in hepatic tissue, or upregulation of extracellular IL-11 or IL-11Ra.

In accordance with the various aspects disclosed herein, in some embodiments the alcoholic liver disease is characterised by one or more of the following (relative to the healthy, non-diseased state): increased serum ALT level; increased liver-to-body weight ratio; reduced bodyweight; increased liver triglyceride level; increased serum IL-11 level; increased gene and/or protein expression of IL-11 in the liver; increased gene and/or protein expression of one or more proinflammatory factors (e.g. selected from TNFa, TIMP1, IL-10, CXCL1 , IL-1 b, and MIP2) in the liver; increased activation of ERK in the liver (i.e. increased the level of pERK in hepatic tissue); increased steatosis of hepatic tissue; increased infiltration of neutrophils (e.g. MPO+ neutrophils) into hepatic tissue; and/or increased infiltration of macrophages (e.g. F4/80+ macrophages) into hepatic tissue. In accordance with the various aspects disclosed herein, in some embodiments alcoholic liver disease is characterised by reduced/impaired liver function relative to function in the absence of the disease.

Treatment may be effective to reduce/delay/prevent the development or progression of alcoholic liver disease. Treatment may be effective to reduce/delay/prevent the worsening of one or more symptoms of alcoholic liver disease. Treatment may be effective to improve one or more symptoms of alcoholic liver disease. Treatment may be effective to reduce the severity of and/or reverse one or more symptoms of alcoholic liver disease. Treatment may be effective to reverse the effects of alcoholic liver disease.

Prevention may refer to prevention of development of alcoholic liver disease, and/or prevention of worsening of alcoholic liver disease, e.g. prevention of progression of alcoholic liver disease, e.g. to a later/chronic stage (e.g. fibrosis and/or cirrhosis).

In some embodiments, the intervention may be aimed at slowing, stopping and/or reversing impairment of liver function associated with alcoholic liver disease.

In accordance with various aspects of the present invention, a method of treating and/or preventing alcoholic liver disease according to the present disclosure may comprise increasing survival of a subject having alcoholic liver disease.

In accordance with various aspects of the present disclosure, methods are provided which are for, or which comprise (e.g. in the context of treatment/prevention of alcoholic liver disease), one or more of the following: reducing serum ALT level; reducing liver-to-body weight ratio; increasing/maintaining bodyweight; reducing liver triglyceride level; reducing serum IL-11 level; reducing gene and/or protein expression of IL-11 in the liver; reducing gene and/or protein expression of one or more proinflammatory factors (e.g. selected from TNFa, TIMP1, IL-10, CXCL1, IL-1 b, and MIP2) in the liver; reducing activation of ERK in the liver (i.e. reducing the level of pERK in hepatic tissue); reducing steatosis of hepatic tissue; reducing infiltration of neutrophils (e.g. MPO+ neutrophils) into hepatic tissue; and/or reducing infiltration of macrophages (e.g. F4/80+ macrophages) into hepatic tissue.

Also provided are agents according to the present disclosure for use in such methods, and the use of agents according to the present disclosure in manufacture of compositions (e.g. medicaments) for use in such methods. It will be appreciated that the methods comprise administering an agent capable of inhibiting IL-11 -mediated signalling to a subject.

Similarly, one or more of the following may be observed in a subject following therapeutic or prophylactic intervention in accordance with the present disclosure (e.g. compared to the level prior to intervention): reduced serum ALT level; reduced liver-to-body weight ratio; increased/maintained bodyweight; reduced liver triglyceride level; reduced serum IL-11 level; reduced gene and/or protein expression of IL-11 in the liver; reduced gene and/or protein expression of one or more proinflammatory factors (e.g. selected from TNFa, TIMP1, IL-10, CXCL1 , IL-1b, and MIP2) in the liver; reduced activation of ERK in the liver (i.e. reducing the level of pERK in hepatic tissue); reduced steatosis of hepatic tissue; reduced infiltration of neutrophils (e.g. MPO+ neutrophils) into hepatic tissue; and/or reduced infiltration of macrophages (e.g. F4/80+ macrophages) into hepatic tissue.

In some embodiments, therapeutic/prophylactic intervention in accordance with the present disclosure may be described as being ‘associated with’ one or more of the effects described in the preceding paragraph. The skilled person is readily able to evaluate such properties using techniques that are routinely practiced in the art.

In some embodiments, treatment in accordance with the present disclosure may be effective to reverse one or more symptoms of alcoholic liver disease. Such treatment may be effective to reverse symptoms even in the case of established, advanced or severe disease/pathology (e.g. fibrosis and/or cirrhosis).

Administration

Administration of an agent capable of inhibiting IL-11 -mediated signalling is preferably in a "therapeutically effective” or “prophylactically effective” amount, this being sufficient to show benefit to the subject.

The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of the disease and the nature of the agent. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disease/condition to be treated, the condition of the individual subject, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington’s Pharmaceutical Sciences, 20th Edition, 2000, pub. Lippincott, Williams & Wlkins.

Multiple doses of the agent may be provided. One or more, or each, of the doses may be accompanied by simultaneous or sequential administration of another therapeutic agent.

Multiple doses may be separated by a predetermined time interval, which may be selected to be one of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or31 days, or 1 , 2, 3, 4, 5, or 6 months. By way of example, doses may be given once every 7, 14, 21 or 28 days (plus or minus 3, 2, or 1 days).

In therapeutic applications, agents capable of inhibiting IL-11 -mediated signalling are preferably formulated as a medicament or pharmaceutical together with one or more other pharmaceutically acceptable ingredients well known to those skilled in the art, including, but not limited to, pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, preservatives, anti-oxidants, lubricants, stabilisers, solubilisers, surfactants (e.g., wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents. The term "pharmaceutically acceptable" as used herein pertains to compounds, ingredients, materials, compositions, dosage forms, etc., which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of the subject in question (e.g., human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, adjuvant, excipient, etc. must also be "acceptable" in the sense of being compatible with the other ingredients of the formulation.

Suitable carriers, adjuvants, excipients, etc. can be found in standard pharmaceutical texts, for example, Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Company, Easton, Pa., 1990; and Handbook of Pharmaceutical Excipients, 2nd edition, 1994.

The formulations may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with a carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with carriers (e.g., liquid carriers, finely divided solid carrier, etc.), and then shaping the product, if necessary.

The formulations may be prepared for suitable administration in accordance with the disease/condition to be treated, e.g. parenteral, systemic, intravenous, intra-arterial, intramuscular, intrathecal, topical, intraocular, intra-conjunctival, subcutaneous, oral ortransdermal routes of administration which may include injection. Injectable formulations may comprise the selected agent in a sterile or isotonic medium. The formulation and mode of administration may be selected according to the agent and disease to be treated.

In some embodiments, agents capable of inhibiting IL-11 -mediated signalling according to the present disclosure may be formulated and/or modified to facilitate delivery to, and/or uptake by, the liver, hepatic tissue, and/or a liver cell (e.g. hepatic stellate cells and/or hepatocytes).

Detection of IL-11 and receptors for IL-11 Some aspects and embodiments of the present invention concern detection of expression of IL-11 or a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130) in a sample obtained from a subject.

In some aspects and embodiments the present invention concerns the upregulation of expression (overexpression) of IL-11 or a receptor for IL-11 (as a protein or oligonucleotide encoding the respective IL-11 or receptor for IL-11) and detection of such upregulation as an indicator of suitability for treatment with an agent capable of inhibiting the action of IL-11 or with an agent capable of preventing or reducing the expression of IL-11 or a receptor for IL-11. Upregulated expression comprises expression at a level that is greater than would normally be expected for a cell or tissue of a given type. Upregulation may be determined by measuring the level of expression of the relevant factor in a cell or tissue. Comparison may be made between the level of expression in a cell or tissue sample from a subject and a reference level of expression for the relevant factor, e.g. a value or range of values representing a normal level of expression of the relevant factor for the same or corresponding cell or tissue type. In some embodiments reference levels may be determined by detecting expression of IL-11 or a receptor for IL-11 in a control sample, e.g. in corresponding cells or tissue from a healthy subject or from healthy tissue of the same subject. In some embodiments reference levels may be obtained from a standard curve or data set. Levels of expression may be quantitated for absolute comparison, or relative comparisons may be made.

In some embodiments upregulation of IL-11 or a receptor for IL-11 (e.g. IL-11 Ra, gp130, ora complex containing IL-11 Ra and/or gp130) may be considered to be present when the level of expression in the test sample is at least 1.1 times that of a reference level. More preferably, the level of expression may be selected from one of at least 1.2, at least 1.3, at least 1.4, at least 1.5, at least 1.6, at least 1.7, at least 1.8, at least 1.9, at least 2.0, at least 2.1 , at least 2.2, at least 2.3, at least 2.4 at least 2.5, at least 2.6, at least 2.7, at least 2.8, at least 2.9, at least 3.0, at least 3.5, at least 4.0, at least 5.0, at least 6.0, at least 7.0, at least 8.0, at least 9.0, or at least 10.0 times that of the reference level.

Expression levels may be determined by one of a number of known in vitro assay techniques, such as PCR based assays, in situ hybridisation assays, flow cytometry assays, immunological or immunohistochemical assays.

By way of example suitable techniques involve a method of detecting the level of IL-11 or a receptor for IL-11 in a sample by contacting the sample with an agent capable of binding IL-11 or a receptor for IL-11 and detecting the formation of a complex of the agent and IL-11 or receptor for IL-11. The agent may be any suitable binding molecule, e.g. an antibody, polypeptide, peptide, oligonucleotide, aptamer or small molecule, and may optionally be labelled to permit detection, e.g. visualisation, of the complexes formed. Suitable labels and means for their detection are well known to those in the art and include fluorescent labels (e.g. fluorescein, rhodamine, eosine and NDB, green fluorescent protein (GFP), chelates of rare earths such as europium (Eu), terbium (Tb) and samarium (Sm), tetramethyl rhodamine, Texas Red, 4- methyl umbelliferone, 7-amino-4-methyl coumarin, Cy3, Cy5), isotope markers, radioisotopes (e.g. 32P, 33P, 35S), chemiluminescence labels (e.g. acridinium ester, luminol, isoluminol), enzymes (e.g. peroxidase, alkaline phosphatase, glucose oxidase, beta-galactosidase, luciferase), antibodies, ligands and receptors. Detection techniques are well known to those of skill in the art and can be selected to correspond with the labelling agent. Suitable techniques include PCR amplification of oligonucleotide tags, mass spectrometry, detection of fluorescence or colour, e.g. upon enzymatic conversion of a substrate by a reporter protein, or detection of radioactivity.

Assays may be configured to quantify the amount of IL-11 or receptor for IL-11 in a sample. Quantified amounts of IL-11 or receptor for IL-11 from a test sample may be compared with reference values, and the comparison used to determine whether the test sample contains an amount of IL-11 or receptor for IL- 11 that is higher or lower than that of the reference value to a selected degree of statistical significance.

Quantification of detected IL-11 or receptor for IL-11 may be used to determine up- or down-regulation or amplification of genes encoding IL-11 or a receptor for IL-11. In cases where the test sample contains fibrotic cells, such up-regulation, down-regulation or amplification may be compared to a reference value to determine whether any statistically significant difference is present.

A sample obtained from a subject may be of any kind. A biological sample may be taken from any tissue or bodily fluid, e.g. a blood sample, blood-derived sample, serum sample, lymph sample, semen sample, saliva sample, synovial fluid sample. A blood-derived sample may be a selected fraction of a patient’s blood, e.g. a selected cell-containing fraction or a plasma or serum fraction. A sample may comprise a tissue sample or biopsy; or cells isolated from a subject. Samples may be collected by known techniques, such as biopsy or needle aspirate. Samples may be stored and/or processed for subsequent determination of IL-11 expression levels.

Samples may be used to determine the upregulation of IL-11 or receptor for IL-11 in the subject from which the sample was taken.

In some preferred embodiments a sample may be a tissue sample, e.g. biopsy, taken from the liver/hepatic tissue. A sample may be obtained from the liver. A sample may comprise hepatic tissue or liver cells.

A subject may be selected for therapy/prophylaxis in accordance with the present invention based on determination that the subject has an upregulated level of expression of IL-11 or of a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130). Upregulated expression of IL-11 or of a receptor for IL-11 may serve as a marker of alcoholic liver disease suitable for treatment with an agent capable of inhibiting IL-11 mediated signalling.

Upregulation may be in a given organ (e.g. the liver), tissue (e.g. hepatic tissue) or in selected cells from a given tissue (e.g. hepatic cells, e.g. hepatic stellate cells and/or hepatocytes). Upregulation of expression of IL-11 or of a receptor for IL-11 may also be determined in a circulating fluid, e.g. blood, or in a blood derived sample. Upregulation may be of extracellular IL-11 or IL-11Ra. In some embodiments expression may be locally or systemically upregulated.

Following selection, a subject may be administered with an agent capable of inhibiting IL-11 mediated signalling.

Diagnosis and prognosis

Detection of upregulation of expression of IL-11 or a receptor for IL-11 (e.g. IL-11 Ra, gp130, or a complex containing IL-11 Ra and/or gp130) may also be used in a method of diagnosing alcoholic liver disease, identifying a subject at risk of developing alcoholic liver disease, and in methods of prognosing or predicting a subject’s response to treatment with an agent capable of inhibiting IL-11 mediated signalling.

“Developing”, “development” and other forms of “develop” may refer to the onset of a disorder/disease, or the continuation or progression of a disorder/disease.

In some embodiments, a subject may be suspected of having or suffering from alcoholic liver disease, e.g. based on the presence of other symptoms indicative of alcoholic liver disease in the subject’s body or in selected cells/tissues of the subject’s body (e.g. the liver/hepatic tissue/liver cells). In some embodiments, a subject may be considered at risk of developing alcoholic liver disease, e.g. because of genetic predisposition; for example, the subject may comprise one or more copies of one or more of the following alleles: PNPLA3 comprising rs738409-G, TM6SF2 comprising rs58542926-T, MBOAT7 comprising rs641738-T, MARC1 comprising rs2642438-C/G/T, and HNRNPUL1 comprising rs15052-C. In some embodiments, a subject may be considered at risk of developing alcoholic liver disease because of exposure to conditions known to be risk factors for alcoholic liver disease, e.g. excessive alcohol consumption (e.g. regular consumption of >40 g ethanol/day for males, >20 g ethanol/day for females).

Determination of upregulation of expression of IL-11 or a receptor for IL-11 may confirm a diagnosis or suspected diagnosis, or may confirm that the subject is at risk of developing alcoholic liver disease. The determination may also diagnose alcoholic liver disease or predisposition as one suitable for treatment with an agent capable of inhibiting IL-11 -mediated signalling.

As such, a method of providing a prognosis for a subject having, or suspected of having alcoholic liver disease may be provided, the method comprising determining whether the expression of IL-11 or a receptor for IL-11 is upregulated in a sample obtained from the subject and, based on the determination, providing a prognosis for treatment of the subject with an agent capable of inhibiting IL-11 -mediated signalling.

In some aspects, methods of diagnosis or methods of prognosing or predicting a subject’s response to treatment with an agent capable of inhibiting IL-11 -mediated signalling may not require determination of the expression of IL-11 or a receptor for IL-11 , but may be based on determining genetic factors in the subject that are predictive of upregulation of expression or activity. Such genetic factors may include the determination of genetic mutations, single nucleotide polymorphisms (SNPs) or gene amplification in IL- 11, IL-11 Ra and/or gp130 which are correlated with and/or predictive of upregulation of expression or activity and/or IL-11 mediated signalling. The use of genetic factors to predict predisposition to a disease state or response to treatment is known in the art, e.g. see Peter Starkel Gut 2008;57:440-442; Wright et al., Mol. Cell. Biol. March 2010 vol. 30 no. 6 1411-1420.

Genetic factors may be assayed by methods known to those of ordinary skill in the art, including PCR based assays, e.g. quantitative PCR, competitive PCR. By determining the presence of genetic factors, e.g. in a sample obtained from a subject, a diagnosis may be confirmed, and/or a subject may be classified as being at risk of developing a disease/condition described herein, and/or a subject may be identified as being suitable for treatment with an agent capable of inhibiting IL-11 mediated signalling.

Some methods may comprise determination of the presence of one or more SNPs linked to secretion of IL-11 or susceptibility to development of alcoholic liver disease. SNPs are usually bi-allelic and therefore can be readily determined using one of a number of conventional assays known to those of skill in the art (e.g. see Anthony J. Brookes. The essence of SNPs. Gene Volume 234, Issue 2, 8 July 1999, 177-186; Fan et al., Highly Parallel SNP Genotyping. Cold Spring Harb Symp Quant Biol 2003. 68: 69-78; Matsuzaki et al., Parallel Genotyping of Over 10,000 SNPs using a one-primer assay on a high-density oligonucleotide array. Genome Res. 2004. 14: 414-425).

The methods may comprise determining which SNP allele is present in a sample obtained from a subject. In some embodiments determining the presence of the minor allele may be associated with increased IL- 11 secretion or susceptibility to development of alcoholic liver disease.

Accordingly, in one aspect of the present invention a method for screening a subject is provided, the method comprising: obtaining a nucleic acid sample from the subject; determining which allele is present in the sample at the polymorphic nucleotide position of one or more of the SNPs listed in Figure 33, Figure 34, or Figure 35 of WO 2017/103108 A1 (incorporated by reference herein), ora SNP in linkage disequilibrium with one of the listed SNPs with an r 2 > 0.8.

The determining step may comprise determining whether the minor allele is present in the sample at the selected polymorphic nucleotide position. It may comprise determining whether 0, 1 or 2 minor alleles are present.

The screening method may be, or form part of, a method for determining susceptibility of the subject to development of alcoholic liver disease, or a method of diagnosis or prognosis as described herein.

The method may further comprise the step of identifying the subject as having susceptibility to, or an increased risk of, developing alcoholic liver disease, e.g. if the subject is determined to have a minor allele at the polymorphic nucleotide position. The method may further comprise the step of selecting the subject for treatment with an agent capable of inhibiting IL-11 mediated signalling and/or administering an agent capable of inhibiting IL-11 mediated signalling to the subject in order to provide a treatment for alcoholic liver disease in the subject or to prevent development or progression of alcoholic liver disease in the subject.

In some embodiments, a method of diagnosing alcoholic liver disease, identifying a subject at risk of developing alcoholic liver disease, and methods of prognosing or predicting a subject’s response to treatment with an agent capable of inhibiting IL-11 mediated signalling employs an indicator that is not detection of upregulation of expression of IL-11 or a receptor for IL-11 , or genetic factors.

In some embodiments, a method of diagnosing alcoholic liver disease, identifying a subject at risk of developing alcoholic liver disease, and methods of prognosing or predicting a subject’s response to treatment with an agent capable of inhibiting IL-11 mediated signalling is based on detecting, measuring and/or identifying one or more indicators of hepatic function and/or damage to hepatic tissue/cells.

Methods of diagnosis or prognosis may be performed in vitro on a sample obtained from a subject, or following processing of a sample obtained from a subject. Once the sample is collected, the patient is not required to be present for the in vitro method of diagnosis or prognosis to be performed and therefore the method may be one which is not practised on the human or animal body. The sample obtained from a subject may be of any kind, as described herein above.

Other diagnostic or prognostic tests may be used in conjunction with those described here to enhance the accuracy of the diagnosis or prognosis or to confirm a result obtained by using the tests described herein.

Subjects

Subjects may be animal or human. Subjects are preferably mammalian, more preferably human. The subject may be a non-human mammal, but is more preferably human. The subject may be male or female. The subject may be a patient.

The patient may have alcoholic liver disease as described herein. A subject may have been diagnosed with alcoholic liver disease, may be suspected of having alcoholic liver disease, or may be at risk from developing alcoholic liver disease.

In embodiments according to the present invention the subject is preferably a human subject. In embodiments according to the present invention, a subject may be selected for treatment according to the methods based on characterisation for certain markers of alcoholic liver disease.

Sequence identity

Pairwise and multiple sequence alignment for the purposes of determining percent identity between two or more amino acid or nucleic acid sequences can be achieved in various ways known to a person of skill in the art, for instance, using publicly available computer software such as ClustalOmega (Soding, J.

2005, Bioinformatics 21, 951-960), T-coffee (Notredame etai. 2000, J. Mol. Biol. (2000) 302, 205-217), Kalign (Lassmann and Sonnhammer 2005, BMC Bioinformatics, 6(298)) and MAFFT (Katoh and Standley 2013, Molecular Biology and Evolution, 30(4) 772-780) software. When using such software, the default parameters, e.g. for gap penalty and extension penalty, are preferably used. Sequences

The invention includes the combination of the aspects and preferred features described except where such a combination is clearly impermissible or expressly avoided.

The features disclosed in the foregoing description, or in the following claims, or in the accompanying drawings, expressed in their specific forms or in terms of a means for performing the disclosed function, or a method or process for obtaining the disclosed results, as appropriate, may, separately, or in any combination of such features, be utilised for realising the invention in diverse forms thereof.

For the avoidance of any doubt, any theoretical explanations provided herein are provided for the purposes of improving the understanding of a reader. The inventors do not wish to be bound by any of these theoretical explanations.

Any section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.

Throughout this specification, including the claims which follow, unless the context requires otherwise, the word “comprise” and “include”, and variations such as “comprises”, “comprising”, and “including” will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Ranges may be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by the use of the antecedent “about,” it will be understood that the particular value forms another embodiment. The term “about” in relation to a numerical value is optional and means for example +/- 10%.

Methods disclosed herein may be performed, or products may be present, in vitro, ex vivo, or in vivo. The term “in vitro" is intended to encompass experiments with materials, biological substances, cells and/or tissues in laboratory conditions or in culture whereas the term “in vivo" is intended to encompass experiments and procedures with intact multi-cellular organisms. In some embodiments, methods performed in vivo may be performed on non-human animals. “Ex vivo" refers to something present or taking place outside an organism, e.g. outside the human or animal body, which may be on tissue (e.g. whole organs) or cells taken from the organism.

Where a nucleic acid sequence is disclosed herein, the reverse complement thereof is also expressly contemplated.

For standard molecular biology techniques, see Sambrook, J., Russel, D.W. Molecular Cloning, A Laboratory Manual. 3 ed. 2001 , Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press

Aspects and embodiments of the present invention will now be discussed with reference to the accompanying figures. Further aspects and embodiments will be apparent to those skilled in the art. All documents mentioned in this text are incorporated herein by reference in their entirety. While the invention has been described in conjunction with the exemplary embodiments described below, many equivalent modifications and variations will be apparent to those skilled in the art when given this disclosure. Accordingly, the exemplary embodiments of the invention set forth above are considered to be illustrative and not limiting. Various changes to the described embodiments may be made without departing from the spirit and scope of the invention.

Brief Description of the Figures

Embodiments and experiments illustrating the principles of the invention will now be discussed with reference to the accompanying figures.

Figures 1 A to 1 E. Schematic, bar charts and graph showing that therapeutic administration anti-IL-11 RA antibody ameliorates liver injury in a mouse model of alcoholic liver disease. (1 A) Schematic timeline of the procedure. C57BL/6J female mice were pair-fed (‘Pair-fed’) or EtOH-fed for 15 days. The EtOH- group received either anti-IL-11RA antibody (‘IL-11RA’) or IgG control (‘IgG’) intraperitoneally. (1B) Administration of EtOH-fed mice with anti-IL-11 RA antibody resulted in significantly lower levels of ALT compared to treatment with IgG control. (1 C, 1 D, 1 E) EtOH-fed mice treated with anti-IL-11 RA antibody displayed (1C) decreased liver-to-body ratio, (1D) decreased weight loss and (1E) decreased hepatic triglyceride accumulation relative to EtOH-fed mice treated with IgG control. *p <0.05, **p <0.01 ; n >5/group. IL-11 RA = anti-lnterleukin 11 Receptor a antibody, EtOH = Ethanol, IgG = IgG isotype-matched control antibody, ALT = alanine transferase, IL-11 = Interleukin 11.

Figures 2A to 2R. Images and bar charts showing that antagonism of IL-11 -mediated signalling reduces the level of IL-11 protein in the liver and ameliorates hepatic inflammation and fibrosis in alcoholic liver disease. (2A) Representative images of hepatic tissue sections harvested from mice in the indicated treatment groups and stained for IL-11. (2B) Administration of anti-IL-11 RA antibody reduced the level of IL-11 in liver tissue of EtOH-fed mice relative to EtOH-fed mice treated with IgG control. (2C to 2H) EtOH- fed mice administered anti-IL-11RA antibody displayed reduced gene expression of the proinflammatory cytokines (2C) TNFa, (2D) TIMP1 , (2E) IL-10, (2F) CXCL1 , (2G) IL-1 b, and (2H) MIP2, relative to EtOH- fed mice administered IgG control. (2I) Representative immunoblot of pERK and ERK in liver tissue from mice in the different treatment groups. (2J) Glycogen score of EtOH-fed mice treated with anti-IL-11 RA antibody trended towards an increased score relative to EtOH-fed mice administered IgG control. (2K to 2R) show gene expression of (2K) Coll A2, (2L) Col3A1 , (2M) Coll A1 , (2N) CCL5, (20) MCP1 , (2P) PPARa, and (2Q and 2R) PPARy in the hepatic tissue of mice of the different treatment groups. In (2Q), the result of analysis of the data by T-test is shown, and in (2R), the result of analysis of the data by ANOVA is shown. *p <0.05, **p <0.01 , ***p <0.001 , ns = non-significant; n >5/group. IL-11 RA = anti- interleukin 11 Receptor a antibody, EtOH = Ethanol, IgG = IgG isotype-matched control antibody, IL-11 = Interleukin 11 , TIMP1 = Tissue Inhibitor of Metalloproteinases 1 , I L- 10 = Interleukin-10, TNFa = Tumor Necrosis Factor a, CXCL-1 = Chemokine (C-X-C motif) Ligand 1 , IL-1 b = Interleukin 1b, MIP2 - Macrophage Inflammatory Protein 2.

Figures 3A to 3F. Images and bar charts showing that antagonism of IL-11 -mediated signalling ameliorates histological inflammation in alcoholic liver disease. (3A and 3B) Representative (3A) images and (3B) quantification of hematoxylin and eosin staining of hepatic tissue sections harvested from mice in the indicated treatment groups. EtOH-fed mice treated with anti-IL-11 RA antibody had a significantly lower hepatic steatosis score compared to EtOH-fed mice administered IgG control. (3C and 3D) Representative (3C) images and (3D) quantification of MPO+ neutrophils in hepatic tissue sections harvested from mice in the indicated treatment groups. EtOH-fed mice treated with anti-IL-11 RA antibody had significantly fewer MPO+ neutrophils compared to EtOH-fed mice administered IgG control. (3E and 3F) Representative (3E) images and (3F) quantification of F4/80+ macrophages in hepatic tissue sections harvested from mice in the indicated treatment groups. EtOH-fed mice treated with anti-IL-11 RA antibody had significantly fewer F4/80+ macrophages compared to EtOH-fed mice administered IgG control. *p <0.05, **p <0.01 , ***p <0.001 , n >5/group. IL-11 RA = anti-l nterleukin 11 Receptor a antibody, EtOH = Ethanol, IgG = IgG isotype-matched control antibody, HPF = high power field, MPO+ = myeloperoxidase positive, F4/80+ = F4/80 positive.

Figures 4A to 4E. Schematic, bar charts and graph showing that therapeutic administration of anti-l L- 11 RA antibody ameliorates liver injury in a mouse model of alcoholic liver disease, when treatment is commenced after EtOH insult. (4A) Schematic timeline of the procedure. C57BL/6J female mice were pair-fed (‘Pair-fed’) or EtOH-fed for 15 days. The EtOH- group received either anti-IL-11 RA antibody (‘IL- 11 RA’) or IgG control (‘IgG’ or ‘IgG Ab’) intraperitoneally from day 7. (4B) Administration of EtOH-fed mice with anti-IL-11 RA antibody resulted in significantly lower levels of ALT compared to treatment with IgG control. (4C, 4D, 4E) EtOH-fed mice treated with anti-IL-11 RA antibody displayed (4C) decreased liver-to-body ratio, (4D) decreased weight loss and (4E) decreased hepatic triglyceride accumulation, relative to EtOH-fed mice treated with IgG control. *p <0.05, ns = non-significant; n >5/group. IL-11RA = anti-l nterleukin 11 Receptor a antibody, EtOH = Ethanol, IgG = IgG isotype-matched control antibody, ALT = alanine transferase, IL-11 = Interleukin 11. Examples

In the following Examples, the inventors demonstrate that IL-11-meidated signalling drives alcohol-related liver disease, and that inhibition of IL-11 -mediated signalling ameliorates symptoms of alcoholic liver disease.

Example 1 : Materials and Methods

Animal Studies

C57BL/6 mice purchased from Jacksons Laboratories (Bar Harbor, ME) were cohoused in the Animal Facility of Medical University of Innsbruck for one week prior to the start of experiments. All mice were fed the Lieber DeCarli pair-fed diet for five days to become acclimated to a liquid diet. Female wild-type (wt) mice (7-8 weeks old) were then fed with a Lieber-DeCarli diet (BioServ, Flemington, NJ) containing an increasing amount of ethanol (EtOH) ranging from 1 to 5%vol ad libitum for 15 days (EtOH-fed) [Bertola et al., Nat Protoc (2013) 8:627-637] Control diet was supplemented with an isocaloric amount of maltose (pair-fed). Pair-fed mice were calorie matched with the ethanol-fed mice. Mice were weighed every other day. 8 hours after gavage the mice were euthanized. All mice received Xylain 5 mg/kg bodyweight (Intervet, Vienna, Austria) and Ketamin 100 mg/kg bodyweight (AniMedica, Senden, Germany) for anesthesia. Blood and tissue samples of liver and intestine were collected afterwards. Serum and tissue samples were stored at -80°C or in RNAIater solution (Qiagen, Hilden, Germany) at -20°C.

In vivo administration of anti-IL-11RA antibody

For antibody treatments, mice were injected intraperitoneally with 20 mg/kg of anti-IL-11 RA antibody X209, or an identical amount of IgG control (11E10, Aldevron; which is produced from 1.10E+11 cells (ATCC, No. CRL-1907)). X209 is a mouse anti-mouse IL-11Ra IgG, and is described e.g. in Widjaja et al., Gastroenterology (2019) 157(3):777-792. X209 is also referred to as “Enx209”, and comprises the VH region according to SEQ ID NO:7 of WO 2019/238884 A1 (SEQ ID NO:32 of the present disclosure), and the VL region according to SEQ ID NO:14 of WO 2019/238884 A1 (SEQ ID NO:33 of the present disclosure).

All anti-IL-11 RA experiments adhered to ethical principles according to Austrian law (BMWFW- 2020.0.547.764) and were carried out at the animal facility of the Medical University of Innsbruck.

ALT analyses

Mouse bodyweights and liver weights were measured. Serum ALT levels were analyzed using an enzymatic assay kit from BQ Kits, Inc. (San Diego, CA) in accordance with the manufacturer ' s instructions.

Triglyceride measurements

For the evaluation of liver triglyceride levels, frozen liver samples were homogenized in PBS. The volume was adjusted to the liver tissue weight. Afterwards, samples were incubated for 30 minutes at 60°C, followed by centrifugation (12,000g, 10 min, room temperature). Supernatants were harvested and triglyceride was isolated in fat-free BSA (Sigma, St Louis, MO)-coated vials. The concentration of triglyceride was measured using TG-reagent (Roche, Basel, Switzerland).

RNA isolation from tissue Tissue RNA was purified by homogenization of samples in TRIzol® reagent (Thermo Fisher Scientific, Waltham, MA). The reverse transcription system (Thermo Fisher Scientific, Waltham, MA) was used to accomplish reverse transcription. Afterwards, qPCR was performed using qPCR SybrGreen (Eurogentec, Seraing, Belgium) and the Mx3000 qPCR cycler (Stratagene, San Diego, CA), and the primers shown in the table below.

Western blot

Hepatic proteins were extracted using the T-PER tissue protein extraction reagent, which was supplemented with HALT proteinase inhibitor cocktail (Thermo Fisher Scientific, Waltham, MA, USA). The protein concentrations were measured by BCA Protein Assay (Pierce, Thermo Fisher Scientific, Waltham, MA, USA) and thereafter separated by SDS-PAGE (Hercules, Bio Rad, CA, USA) and blotted onto Hybond-P PVDF membranes (GE Healthcare, Chicago, IL, USA). The SNAP i.d.® protein detection system (Millipore, Burlington, MA, USA) was used for blocking, washing, and ERK and pERK incubation. Immunoreactivity was visualized by using chemiluminescens on Amersham Hyperfilms (GE Healthcare, Chicago, IL, USA). GAPDH (glyceraldehyde-3-phosphate dehydrogenase) served as reference protein. Western blot signals were quantitated with Biorad ChemiDoc MP imaging system (Hercules, CA, USA).

Histology

For histological analysis, liver sections were stained with hematoxylin and eosin (H&E), or were stained with antibodies specific for IL-11 , myeloperoxidase or F4/80. A pathologist analyzed the H&E- IL-11-, myeloperoxidase- and F4/80-stained liver sections in a blinded fashion with regard to hepatic steatosis, inflammation, infiltration of inflammatory cells and positive cells for IL-11 , myeloperoxidase and F4/80. Hepatic steatosis was quantified as percentage of cells showing lipid accumulation, with a maximum steatosis score of 300. Glycogen was analyzed as relative content in liver tissue by an independent pathologist as well.

IL-11 immunohistochemistry

Formalin-fixed paraffin-embedded sections were de-paraffinised and rehydrated. Slides were peroxidase blocked. The primary antibody (anti-IL-11, diluted 1:200; R&D systems, Minneapolis, MN) was incubated overnight at 4°C. Secondary biotinylated antibody (Vector, DAKO, Santa Clara, CA) was incubated for one hour at room temperature. The antibody incubation steps were done in a chamber in a humidified atmosphere. Immune-reactivity was visualized with horseradish peroxidase (HRP)-driven 3,3'- diaminobenzidine (DAKO, Santa Clara, CA). Stained sections were scanned and analyzed by an expert pathologist.

Myeloperoxidase immunohistochemistry

Mouse liver sections were d e paraffin ated in xylene and dehydrated in an ethanol gradient. Antigen was unmasked with 2% citrate buffer (pH=6; Vector Laboratories, Burlingame, CA) in a conventional steamer. Endogenous peroxidase activity was blocked with peroxidase (Dako, Santa Clara, CA) and protein blocking was performed using a ready-to-use kit (MP-740, Dako, Santa Clara, CA). Rabbit antimyeloperoxidase (Dako, Santa Clara, CA) and secondary anti-rabbit antibodies (Vector Laboratories, Burlingame, CA) served to visualize myeloperoxidase. Tissue samples were stained with DAB (Dako, Santa Clara, CA) and counterstained with hematoxylin (Dako, Santa Clara, CA). MPO positive cells were counted in ten randomly selected high-power fields by a technician in a blinded manner.

F4/80 immunohistochemistry

A specific anti-F4/80 rabbit monoclonal antibody (CellSignaling, Cambridge, UK) and a second anti-rabbit antibody (Vector Laboratories, Burlingame, CA) stained F4/80. Immunoreactivity was visualize with DAB and samples were counterstained with hematoxylin (Dako, Santa Clara, CA). F4/80 positive cells were counted in ten randomly selected high-power fields by technician in a blinded manner.

Data are expressed as mean ± standard error of mean or as median with first and third quartiles. For comparing quantitative variables, the Student’s t-test or the non-parametric Mann-Whitney U or Wilcoxon signed-rank test or ANOVA were used, as appropriate. Normality of distribution was determined by Kolmogorov-Smirnovtest. The correlation analysis was estimated using the Spearman’s p coefficient. Outliers were identified using the ROUT method or Grubbs’ test. A p-value < 0.05 was considered as statistically significant. All statistical analyses were performed using SPSS Statistics v.22 (IBM, Chicago, IL) and GraphPad PRISM 5 (La Jolla, CA).

Example 2: Results

Inhibition

Female C57BL/6J mice were fed a 5% ethanol containing Lieber-DeCarli diet or an isocaloric pair diet for 15 days. In this experimental set-up EtOH-fed mice received antagonist anti-IL-11 RA antibody X209 or IgG control intraperitoneally as illustrated in Figure 1A. Control IgG-treated EtOH-fed mice showed signs of liver injury which was reversed by anti-IL-11 RA antibody administration (Figure 1 B). Furthermore, anti- IL-11RA antibody administration resulted in reduced liver-body ratio compared to IgG control (Figure 1C) and anti-IL-11RA antibody also prevented EtOH induced weight loss (Figure 1D). Treatment with anti-IL- 11 RA antibody also inhibited the EtOH-induced accumulation of hepatic triglyceride observed in IgG control-treated EtOH-fed mice (Figure 1E).

Inhibition of I L-11 -mediated signaling protects against alcohol-induced liver inflammation EtOH-fed mice treated with anti-IL-11 RA antibody were found to have significantly lower expression of II- 11 protein in hepatic tissue compared to EtOH-fed mice administered IgG control (Figures 2A and 2B). The inventors investigated whether upregulated expression of IL-11 in the livers of EtOH-fed mice upregulated the expression of other pro-inflammatory mediators, and found that hepatic gene expression ofTNFa, tissue inhibitor of metalloproteinases 1 ( Timpl ), 1110, Cxcl1, II1b, and macrophage inflammatory protein 2 ( Mip2 ) was upregulated in EtOH-fed mice administered IgG control (Figures 2C to 2H). Treatment with anti-IL-11 RA antibody was found to significantly reduce gene expression of each of these pro-inflammatory mediators. Figures 2J to 2R show the results of analysis of several other markers of fibrosis and inflammation.

Liver protection following treatment with anti-IL-11 RA antibody was associated with reduced pErk activation (Figure 2I), in agreement with findings in a model of non-alcoholic fatty liver disease (NAFLD) in mice [Widjaja et al., Gastroenterology (2019) 157:777-792. e714], where IL-11-driven ERK phosphorylation was found to be of central importance for hepatic stellate cell (HSC) transformation and fibrosis.

IL-11RA treatment reduces infiltration of pro-inflammatory cells into the liver

After hematoxylin and eosin-staining of liver tissue, 20 high power fields (HPFs) were analyzed and a steatosis score was calculated. Steatosis score of EtOH-fed mice was significantly reduced upon treatment with anti-IL-11 RA antibody compared to IgG control-treated animals (Figures 3A and 3B).

Myeloperoxidase (MPO) staining demonstrated that anti-IL-11 RA antibody treatment strongly inhibited neutrophil infiltration into liver tissue (Figures 3C and 3D), and significantly fewer F4/80-positive macrophages were observed in the hepatic tissue of EtOH-fed mice treated with anti-IL-11RA antibody, compared to those treated with IgG control (Figures 3E and 3F). Inhibition of I L-11 -mediated signaling reduces experimental ALD in a therapeutic model Female C57BL/6J mice were fed a 5% ethanol containing Lieber-DeCarli diet or an isocaloric pair diet for 15 days. In this experimental set-up EtOH-fed mice received antagonist anti-IL-11 RA antibody X209 or IgG control intraperitoneally from Day 7 as illustrated in Figure 4A. Control IgG-treated EtOH-fed mice showed signs of liver injury which was reversed by anti-IL-11 RA antibody administration (Figure 4B). Furthermore, anti-IL-11RA antibody administration resulted in reduced liver-body ratio compared to IgG control (Figure 4C) and anti-IL-11RA antibody also reduced EtOH induced weight loss (Figure 4D). Treatment with anti-IL-11 RA antibody also inhibited the EtOH-induced accumulation of hepatic triglyceride observed in IgG control-treated EtOH-fed mice (Figure 4E).

Example 3: Discussion

The inventors hypothesised that IL-11 might play an important role in the pro-inflammatory processes in stromal immunity, and that inhibiting IL-11 -mediated signalling via treatment with neutralising antibody to the IL-11 receptor might have beneficial effects on inflammation and correlates of pathology in alcoholic liver disease.

Parenchymal infiltration of neutrophils and macrophages is a prominent feature of alcoholic liver disease, and is likely due to ethanol-mediated activation of innate immunity and subsequent induction of proinflammatory cytokines and chemokines [Gao etai., Gastroenterology (2011) 141:1572-1585; Mandrekaref a/., Hepatology (2016) 64:1343-1355; Seitz eta!., Nat RevDis Primers (2018) 4:16; Louvet et ai., Nat Rev Gastroenterol Hepatol (2015) 12:231-242] Hepatocytes express the IL-11 receptor and secrete cytokines upon ligation, such as transforming growth factor beta (TGFpl). IL-11 -mediated activation of hepatocytes is unexpectedly cytotoxic, and an autocrine and maladaptive loop of IL-11 activity in hepatocytes was apparent in the present model of alcoholic liver disease.

Administration of anti-IL-11 RA antibody was found to robustly reduce inflammation in alcoholic liver disease. The results described hereinabove demonstrate that antagonism of IL-11 -mediated signalling (e.g. using an antibody antagonist of IL-11 -mediated signalling) is effective for the treatment/prevention of alcohol-induced liver disease, and particularly alcoholic hepatitis.